Language selection

Search

Patent 2994555 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2994555
(54) English Title: ANTI-TIGIT ANTIBODIES
(54) French Title: ANTICORPS ANTI-TIGIT
Status: Pre-Grant
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • A61P 35/00 (2006.01)
  • A61P 37/02 (2006.01)
  • C07K 14/705 (2006.01)
  • C07K 16/00 (2006.01)
  • C07K 16/18 (2006.01)
  • C07K 16/28 (2006.01)
  • C07K 16/30 (2006.01)
  • G01N 33/566 (2006.01)
(72) Inventors :
  • WILLIAMS, SYBIL M. G. (United States of America)
  • SEGHEZZI, WOLFGANG (United States of America)
  • FAYADAT-DILMAN, LAURENCE (United States of America)
  • LIANG, LINDA (United States of America)
  • JUAN, VERONICA (United States of America)
(73) Owners :
  • MERCK SHARP & DOHME LLC (United States of America)
(71) Applicants :
  • MERCK SHARP & DOHME CORP. (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2016-08-09
(87) Open to Public Inspection: 2017-02-23
Examination requested: 2021-05-27
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2016/046100
(87) International Publication Number: WO2017/030823
(85) National Entry: 2018-02-01

(30) Application Priority Data:
Application No. Country/Territory Date
62/205,048 United States of America 2015-08-14

Abstracts

English Abstract

The present invention relates to anti-TIGIT antibodies, as well as use of these antibodies in the treatment of diseases such as cancer and infectious disease.


French Abstract

La présente invention concerne des anticorps anti-TIGIT, ainsi que l'utilisation de ces anticorps dans le traitement de maladies telles que le cancer et les maladies infectieuses.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. An antibody or antigen binding fragment thereof that binds to human
TIGIT, wherein the
antibody or antigen binding fragment is selected from the group consisting of:
a an antibody or antigen binding fragment comprising: a heavy chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO:1, a heavy
chain variable region CDR2 comprising the amino acid sequence of SEQ ID
NO:2, a heavy chain variable region CDR3 comprising the amino acid sequence
of SEQ ID NO:3, 45, 46, or 47, a light chain variable region CDR1 comprising
the
amino acid sequence of SEQ ID NO:4, a light chain variable region CDR2
comprising the amino acid sequence of SEQ ID NO:5, and a light chain variable
region CDR3 comprising the amino acid sequence of SEQ ID NO:6, 57, 58, 59,
60, 61, or 62; and
b. an antibody or antigen binding fragment comprising: a heavy chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO:17, a heavy
chain variable region CDR2 comprising the amino acid sequence of SEQ ID
NO:18, 81, 82, 83, 84, 85, 86, 87, or 88, a heavy chain variable region CDR3
comprising the amino acid sequence of SEQ ID NO:19, a light chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO:20, a light chain
variable region CDR2 comprising the amino acid sequence of SEQ ID NO:21, and
a light chain variable region CDR3 comprising the amino acid sequence of SEQ
ID NO:22.
2. The antibody or antigen binding fragment of claim 1, wherein the antibody
comprises a
heavy chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO:1,
a heavy chain variable region CDR2 comprising the amino acid sequence of SEQ
ID
NO:2, a heavy chain variable region CDR3 comprising the amino acid sequence of
SEQ
ID NO:3, 45, 46, or 47, a light chain variable region CDR1 comprising the
amino acid
sequence of SEQ ID NO:4, a light chain variable region CDR2 comprising the
amino acid
sequence of SEQ ID NO:5, and a light chain variable region CDR3 comprising the
amino
acid sequence of SEQ ID NO:6, 57, 58, 59, 60, 61, or 62.
3. The antibody or antigen binding fragment of claim 1, wherein the antibody
comprises a
heavy chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO:17, a heavy chain variable region CDR2 comprising the amino acid sequence
of SEQ
-99-

ID NO: 18, 81, 82, 83, 84, 85, 86, 87, or 88, a heavy chain variable region
CDR3
comprising the amino acid sequence of SEQ ID NO:19, a light chain variable
region
CDR1 comprising the amino acid sequence of SEQ ID NO:20, a light chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO:21, and a light
chain
variable region CDR3 comprising the amino acid sequence of SEQ ID NO:22.
4. An antibody or antigen binding fragment thereof that binds to human MIT
comprising a
light chain immunoglobulin, a heavy chain immunoglobulin or both a light chain
and a
heavy chain immunoglobulin selected from the group consisting of
a. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising the amino acid sequence of SEQ ID NO:7 and/or a variable
light
chain comprising the amino acid sequence of SEQ ID NO:8;
b. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising the amino acid sequence of SEQ ID NO:9 and/or a variable
light
chain comprising the amino acid sequence of SEQ ID NO:13;
c. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising the amino acid sequence of SEQ ID NO: 23 and/or a variable
light chain comprising the amino acid sequence of SEQ ID NO:24;
d. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising the amino acid sequence of SEQ ID NO: 25 and/or a variable
light chain comprising the amino acid sequence of SEQ ID NO:29;
e. an antibody or antigen binding fragment thereof comprising a variable heavy

chain selected from the group consisting of SEQ ID NOs: 10, 11, 12, 48, 49,
50,
51, 52, 53, 54, 55, or 56and/or a variable light chain selected from the group

consisting of any one of SEQ ID NOs: 14, 15, 16, 63, 64, 65, 66, 67, 68, 69,
70,
71, 72, 73, 74, 75, 76, 77, 78, 79, or 80;
f. an antibody or antigen binding fragment thereof comprising a variable heavy

chain selected from the group consisting of SEQ ID NOs: 26, 27, 28, 89, 90,
91,
92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108,
109,
110, 111, and 112and/or a variable light chain selected from the group
consisting
of any one of SEQ ID NOs: 30, 31, and 32;
g. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity any one of
SEQ ID NOs: 10, 11, 12, 48, 49, 50, 51, 52, 53, 54, 55, or 56and/or a variable

light chain comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity to any
-100-

one of SEQ ID NOs: 14, 15, 16, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74,
75,
76, 77, 78, 79, or 80;
h. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity any one of
SEQ ID NOs: 26, 27, 28, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101,
102,
103, 104, 105, 106, 107, 108, 109, 110, 111, or 112 and/or a variable light
chain
comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity to any one of SEQ
ID NOs: 30, 31, or 32;
i. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity any one of
SEQ ID NOs: 10, 11, 12, 48, 49, 50, 51, 52, 53, 54, 55, or 56 and/or a
variable
light chain comprising at least 90%, 95%, 95%, 96%, 97%, 98% or 99% identity
to any one of SEQ ID NOs: 14, 15, 16, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72,
73,
74, 75, 76, 77, 78, 79, or 80, wherein any sequence variations occur in the
framework regions of the antibody; and
j. an antibody or antigen binding fragment thereof comprising a variable heavy

chain comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity any one of
SEQ ID NOs: 26, 27, 28, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101,
102,
103, 104, 105, 106, 107, 108, 109, 110, 111, or 112 and/or a variable light
chain
comprising at least 90%, 95%, 95%, 96%, 97%, 98% or 99% identity to any one
of SEQ ID NOs: 30, 32, or 32, wherein any sequence variations occur in the
framework regions of the antibody.
5. The antibody or antigen binding fragment of any one of claims 1-5, wherein
the antibody
binds to human TIGIT with a KD value of about 1 x 10 -9 M to about 1 x 10 -12
M as
determined by surface plasmon resonance (e.g., BIACORE) or a similar technique
(e.g.
KinExa or OCTET.
6. The antibody or antigen binding fragment of any of the above claims, which
is a
humanized antibody comprising two heavy chains and two light chains.
7. The antibody or antigen binding fragment of any of the above claims, which
is a
humanized antibody, wherein the antibody comprises a human IgGl constant
domain and
a human kappa constant domain.
8. The antibody or antigen binding fragment of any one of the above claims,
which is an
antibody, wherein the antibody is produced in a CHO cell.
-101-

9. An isolated polypeptide comprising the amino acid sequence of any one of
SEQ ID NOs:
7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32,
48, 49, 50, 51, 52,
53, 54, 55, 56, 63, 54, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,
78, 79, 80, 89, 90,
91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107,
108, 109, 110,
111, or 112.
10. An isolated nucleic acid encoding: any one of the antibodies or antigen
binding fragments
of claims 1-8, or any one of the polypeptides of claim 9.
11. An expression vector comprising the isolated nucleic acid of claim 10.
12. A host cell comprising the antibody, binding fragment, polypeptide,
polynucleotide or
expression vector of any of claims 1-11.
13. The host cell of claim 12, which is a Pichia cell or a Chinese hamster
ovary cell.
14. A composition comprising the antibody or antigen binding fragment of any
one of claims
1-8 and a pharmaceutically acceptable carrier or diluent.
15. The composition of claim 14, further comprising an agent selected from the
group
consisting of:
a. an anti-PD 1 antibody or an antigen binding fragment thereof;
b. an anti-LAG3 antibody or an antigen binding fragment thereof;
c. an anti-VISTA antibody or an antigen binding fragment thereof;
d. an anti-BTLA antibody or an antigen binding fragment thereof;
e. an anti-TIM3 antibody or an antigen binding fragment thereof;
f. an anti-CTLA4 antibody or an antigen binding fragment thereof;
g. an anti-HVEM antibody or an antigen binding fragment thereof;
h. an anti-CD27 antibody or an antigen binding fragment thereof;
i. an anti-CD137 antibody or an antigen binding fragment thereof;
j. an anti-OX40 antibody or an antigen binding fragment thereof;
k. an anti-CD28 antibody or an antigen binding fragment thereof;
l. an anti-PDL1 antibody or an antigen binding fragment thereof;
m. an anti-PDL2 antibody or an antigen binding fragment thereof;
n. an anti-GITR antibody or an antigen binding fragment thereof;
o. an anti-ICOS antibody or an antigen binding fragment thereof ;
p. an anti-SIRP.alpha. antibody or an antigen binding fragment thereof;
q. an anti-ILT2 antibody or an antigen binding fragment thereof;
r. an anti-ILT3 antibody or an antigen binding fragment thereof;
s. an anti-ILT4 antibody or an antigen binding fragment thereof; and
-102-

t. an anti-ILT5 antibody or an antigen binding fragment thereof
16. The composition of claim 15, wherein the anti-PD1 antibody or an antigen
binding
fragment thereof is selected from the group consisting of: pembrolizumab or an
antigen
binding fragment thereof and nivolumab or an antigen binding fragment thereof.
17. A method of producing an antibody or antigen binding fragment comprising:
a. culturing a host cell comprising a polynucleotide encoding the heavy chain
and/or
the light chain of any one of the antibodies or antigen binding fragments of
claims
1-8 under conditions favorable to expression of the polynucleotide; and
b. optionally, recovering the antibody or antigen binding fragment from the
host cell
and/or culture medium.
18. A method of treating cancer in a human subject, comprising administering
to the subject
an effective amount of the antibody or antigen binding fragment of any one of
claims 1-8,
optionally in association with a further therapeutic agent or therapeutic
procedure.
19. A method of treating an infection or infectious disease in a human
subject, comprising
administering to the subject an effective amount of the antibody or antigen
binding
fragment of any one of claims 1-8, optionally in association with a further
therapeutic
agent or therapeutic procedure.
20. A vaccine comprising the antibody or antigen binding fragment of any one
of claims 1-8
and an antigen.
21. A method for detecting the presence of a TIGIT peptide or a fragment
thereof in a
sample comprising contacting the sample with an antibody or fragment of any of
claims
1-8 and detecting the presence of a complex between the antibody or fragment
and the
peptide; wherein detection of the complex indicates the presence of the TIGIT
peptide.
22. A method of increasing the activity of an immune cell, comprising
contacting the immune
cell with any one of the antibodies or antigen binding fragments of claims 1-
8.
23. A method of increasing the activity of an immune cell, comprising
administering to a
subject in need thereof an effective amount of an antibody or antigen binding
fragment
according to any one of claims 1-8.
24. The method of claim 23, wherein said method is used for:
a, the treatment of cancer;
b. the treatment of an infection or infectious disease; or
c. as a vaccine adjuvant.
25. An antibody or antigen binding fragment according to any one of claims 1-
8, for use in
the preparation of a medicament to:
-103-

a increase immune cell activation;
b. treat cancer; or
c. treat an infection or infectious disease.
26. Use of the antibody or antigen binding fragment of claims 1-8 for the
manufacture of a
medicament for the treatment of cancer for: increasing immune cell activation;
treating
cancer; or treating an infection or infectious disease.
27. An antibody or antigen binding fragment that binds to an epitope on human
T1GIT
comprising the amino acid sequences SSTTAQVNWEQQDQL (SEQ ID NO:113), ICN,
IYHTYPGT (SEQ ID NO:114), and GRIFL (SEQ ID NO:115).
-104-

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
ANTI-TIGIT ANTIBODIES
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims benefit of U.S. Provisional Application No.
62/205,048 filed August 14, 2015, and which is incoporated herein in its
entirety.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
The sequence listing of the present application is submitted electronically
via
EFS-Web as an ASCII formatted sequence listing with a file name
"24178W0PCTSEQ.txt",
creation date of July 29, 2016, and a size of 111 KB. This sequence listing
submitted via
EFS-Web is part of the specification and is herein incorporated by reference
in its entirety.
FIELD OF THE INVENTION
The present invention relates to anti-T1GIT antibodies, as well as use of
these
antibodies in the treatment of diseases such as cancer and infectious disease.
BACKGROUND OF THE INVENTION
A key factor for enabling tumor irrununotherapy emerged from discoveries that
inhibitory immune modulatory receptors (IMRs), that generally function as
immune
checkpoints to maintain self-tolerance, are central to the ability of tumor
microenvironments
to evade immunity. Blockade of inhibitoty IMRs appears to unleash potent tumor-
specific
immune responses more effectively than direct stimulation of tumor-immunity
with
activating cytokines or tumor vaccines, and this approach has the potential to
transform
human cancer therapy. An important implication and opportunity now arises for
the potential
to develop new antibody antagonists for other 1MRs and to combine antagonist
antibodies to
more than one IMR in order to increase the proportion of responders in
oncology clinical
trials, as well as, expand upon oncology indications in which tumor
immunotherapy
treatments are effective.
Significantly, inhibitory IMRs and ligands that regulate cellular immunity are

commonly overexpressed on tumor cells and tumor associated macrophages (TAMs).
Notably, overexpression of PD-L1 in tumors is associated with tumor specific T
cell
exhaustion and a poor prognosis. Blockade of PD-1/PD-L1 ligation in clinical
trials resulted
in durable tumor regression responses in a substantial proportion of patients.
A recent report
demonstrated that co-expression of PD-1 and another inhibitory IMR (TIM-3) in
melanoma
-1-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
patient derived tumor-specific CD8+ T cells was associated with more
dysfunctional T cell
exhaustion phenotypes compared to cells expressing either TMR alone. Moreover,
several
reports using pre-clinical tumor models demonstrated blockade of multiple
IMRs, including
PD-1, T1M-3, LAG-3 and CTLA-4 more effectively induced anti-tumor responses
than
antagonizing PD-1 alone. These results underscore the importance of further
investigating
IMR pathways.
TIGIT (T cell immunoreceptor with Ig and ITIM domains) is an
irrununomodulatory
receptor expressed primarily on activated T cells and NK cells. TIGIT is also
known as
VSIG9; VSTM3; and WUCAM. Its structure shows one extracellular immunoglobulin
domain, a type 1 transmembrane region and two ITIM motifs. TIGIT forms part of
a co-
stimulatory network that consists of positive (CD226) and negative (TIGIT)
immunomodulatory receptors on T cells, and ligands expressed on APCs (CD155
and
CD112).
An important feature in the structure of TIGIT is the presence of an
immunoreceptor
tyrosine-based inhibition motif (ITIM) in its cytoplasmic tail domain. As with
PD-1 and
C'TLA-4, the MM domain in the cytoplasmic region of TIGIT is predicted to
recruit tyrosine
phosphatases, such as SHP-1 and SHP-2, and subsequent de-phosphoiylation of
tyrosine
residues with in the immunoreceptor tyrosine-base activation motifs (ITAM) on
T cell
receptor (TCR) subunits. Hence, ligation of TIGIT by receptor-ligands CD155
and CD112
expressed by tumor cells or TAMS may contribute to the suppression of TCR-
signaling and T
cell activation, which is essential for mounting effective anti-tumor
immunity. Thus, an
antagonist antibody specific for TIGIT could inhibit the CD155 and CD112
induced
suppression of T cell responses and enhance anti-tumor immunity. It is an
object of the
present invention to obtain an anti-TIGIT antibody that can be used for the
treatment of
cancer, either alone or in combination with other reagents.
SUMMARY OF THE INVENTION
The invention provides anti-TIGIT antibodies and antigen binding fragments
thereof
comprising the structural and functional features specified below.
In one embodiment, the invention provides an antibody or antigen binding
fragment
thereof that binds to human TIGIT, comprising: a heavy chain variable region
CDR3
comprising the amino acid sequence of SEQ ID NO: 3, 19, 45, 46, or 47. In a
further
embodiment, the antibody or antigen binding fragment thereof optionally has at
least one of
the following characteristics: (i) binds to human TIGIT with a KD value of
about 1 x 10-9M
-2-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
to about 1 x 1042 M as determined by surface plasmon resonance (e.g., BIACORE)
or a
similar technique (e.g. KinExa or OCTET); (ii) cross-reacts with cynomolgous
and rhesus
TIGIT; (iii) blocks binding of human TIGIT to human CD155 and human CD112;
(iv)
increases T cell activation: (v) stimulates antigen-specific T-cell production
of IL-2 and
IFNy; (vi) blocks induction of T cell suppression of activation induced by
TIGIT ligation
with cognate ligands CD155 and CD112.
hi another embodiment, the invention provides an antibody or antigen binding
fragment thereof that binds to human TIGIT, comprising: a light chain variable
region CDR3
comprising the amino acid sequence of SEQ ID NO: 6, 22, 57, 58, 59, 60, 61, or
62. In a
further embodiment, the antibody optionally has at least one of the following
characteristics:
(i) binds to human TIGIT with a KD value of about 1 x i0 M to about 1 x 1042 M
as
determined by surface plasmon resonance (e.g., BIACORE) or a similar technique
(e.g.
KinExa or OCTET); (ii) cross-reacts with cynomolgous and rhesus TIGIT; (iii)
blocks
binding of human TIGIT to human CD155 and human CD112; (iv) increases T cell
activation; (v) stimulates antigen-specific T-cell production of IL-2 and
IFNy; (vi) blocks
induction of T cell suppression of activation induced by TIGIT ligation with
cognate ligands
CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
Iraginent thereof that binds to human TIGIT comprising: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; and (iii) a
heavy chain
variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45,
46, or 47.
In a further embodiment, the antibody or antigen binding fragment thereof
optionally has at
least one of the following characteristics: (i) binds to human TIGIT with a KD
value of about
1 x 104 M to about 1 x 1042 M as determined by surface plasmon resonance
(e.g.,
BIACORE) or a similar technique (e.g. KinExa or OCTET); (ii) cross-reacts with
cynomolgous and rhesus -MIT; (iii) blocks binding of human MIT to human CD155
and
human CD112; (iv) increases T cell activation; (v) stimulates antigen-specific
T-cell
production of IL-2 and IFNy; (vi) blocks induction of T cell suppression of
activation induced
by TIGIT ligation with cognate ligands CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a heavy chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85,
86, 87, or
-3-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
88; and (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of SEQ
ID NO: 19. In a further embodiment, the antibody or antigen binding fragment
thereof
optionally has at least one of the following characteristics: (i) binds to
human TIGIT with a
KD value of about 1 x i0 M to about 1 x 10-12 M as determined by surface
plasmon
resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or OCTET); (ii)
cross-reacts
with cynomolgous and rhesus -MIT; (iii) blocks binding of human TIGIT to human
CD155
and human CD112; (iv) increases T cell activation; (v) stimulates antigen-
specific T-cell
production of IL-2 and IFNT; (vi) blocks induction of T cell suppression of
activation induced
by TIGIT ligation with cognate ligands CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a light chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 4; (ii) a light chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 5; and (iii) alight
chain variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 6, 57, 58, 59,
60, 61, or
62. In a further embodiment, the antibody or antigen binding fragment thereof
optionally has
at least one of the following characteristics: (i) binds to human TIGIT with a
KD value of
about 1 x 10-9M to about 1 x 10-12 M as determined by surface plasmon
resonance (e.g.,
BIACORE) or a similar technique (e.g. KinExa or OCTET); (ii) cross-reacts with

cynomolgous and rhesus TIGIT; (iii) blocks binding of human TIGIT to human
CD155 and
human CD112; (iv) increases T cell activation; (v) stimulates antigen-specific
T-cell
production of IL-2 and IFNT; (vi) blocks induction of T cell suppression of
activation induced
by TIGIT ligation with cognate ligands CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a light chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 20; (ii) a light chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 21; and (iii) a light
chain variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 22. In a further
embodiment, the antibody or antigen binding fragment thereof optionally has at
least one of
the following characteristics: (i) binds to human TIGIT with a KD value of
about 1 x 10-9 M
to about 1 x 10-12 M as determined by surface plasmon resonance (e.g.,
BIACORE) or a
similar technique (e.g. KinExa or OCTET); (ii) cross-reacts with cynomolgous
and rhesus
TIGIT; (iii) blocks binding of human TIGIT to human CD155 and human CD112;
(iv)
increases T cell activation; (v) stimulates antigen-specific T-cell production
of IL-2 and
-4-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
IFNT; (vi) blocks induction of T cell suppression of activation induced by
TIGIT ligation
with cognate ligands CD 155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a heavy chain variable
region CDRI
comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain
variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47;
(iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 4; (v)
a light chain variable region CDR2 comprising the amino acid sequence of SEQ
ID NO: 5;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the antibody or antigen
binding
fragment thereof is humanized. In another embodiment, the antibody or antigen
binding
fragment thereof comprises a heavy chain variable region having the amino acid
sequence of
SEQ ID NO: 9; and a light chain variable region having the amino acid sequence
of SEQ ID
NO: 13. In one embodiment, the antibody or antigen binding fragment thereof
comprises a
heavy chain variable region selected from the group consisting of SEQ ID NOs:
10, 11, 12,
48, 49, 50, 51, 52, 53, 54, 55, and 56; and a light chain variable region
selected from the
group consisting of SEQ ID NOs: 14, 15, 16, 63, 64, 65, 66, 67, 68, 69, 70,
71, 72, 73, 74, 75,
76, 77, 78, 79, and 80. In one embodiment, the antibody or antigen binding
fragment thereof
comprises a heavy chain variable region comprising at least 90%, 95%, 96%,
97%, 98% or
99% identity to any one of SEQ ID NOs:10, 11, 12, 48, 49, 50, 51, 52, 53, 54,
55, or 56; and
a light chain variable region comprising at least 90%, 95%, 96%, 97%, 98% or
99% identity
to any one of SEQ ID NOs: 14, 15, 16, 63, 64,65, 66,67, 68, 69, 70, 71, 72,
73, 74, 75, 76,
77, 78, 79, or 80. In one embodiment, the antibody or antigen binding fragment
thereof
comprises a heavy chain variable region comprising at least 90%, 95%, 96%,
97%, 98% or
99% identity to any one of SEQ ID NOs: 10, 11, 12, 48, 49, 50, 51, 52, 53, 54,
55, or 56; and
a light chain variable region comprising at least 90%, 95%, 96%, 97%, 98% or
99% identity
to any one of SEQ ID NOs: 14, 15, 16, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72,
73, 74, 75, 76,
77, 78, 79, or 80, wherein any sequence variations occur in the framework
regions of the
antibody. In a further embodiment of the aforementioned, the antibody or
antigen binding
fragment thereof optionally has at least one of the following characteristics:
(i) binds to
human TIGIT with a KD value of about 1 x 104 M to about 1 x 10-12 M as
determined by
surface plasmon resonance (e.g., BIACORE) or a similar technique (e.g. KinExa
or OCTET);
(ii) cross-reacts with cynomolgous and rhesus TIGIT; (iii) blocks binding of
human TIGIT to
-5-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
human CD155 and human CD112; (iv) increases T cell activation; (v) stimulates
antigen-
specific T-cell production of TL-2 and IFNy; (vi) blocks induction of T cell
suppression of
activation induced by TIGIT ligation with cognate ligands CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a heavy chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85,
86, 87, or
88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of SEQ ID
NO: 19; (iv) alight chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 20; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 21; and (vi) a light chain variable region CDR3 comprising the
amino acid
sequence of SEQ ID NO: 22. In one embodiment, the antibody or antigen binding
fragment
thereof is humanized. In another embodiment, the antibody or antigen binding
fragment
thereof comprises a heavy chain variable region having the amino acid sequence
of SEQ ID
NO:25; and a light chain variable region having the amino acid sequence of SEQ
ID NO:29.
In a further embodiment, the antibody or antigen binding fragment thereof
comprises a heavy
chain variable region selected from the group consisting of SEQ ID NOs: 26,
27, 28, 89, 90,
91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107,
108, 109, 110, 111,
and 112; and a light chain variable region selected from the group consisting
of SEQ ID
NOs:30, 31, and 32. In a further embodiment of the aforementioned, the
antibody or antigen
binding fragment thereof comprises a heavy chain variable region comprising at
least 90%,
95%, 96%, 97%, 98% or 99% identity to any one of SEQ ID NOs: 26, 27, 28, 89,
90, 91, 92,
93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109,
110, 111, or 112;
and a light chain variable region comprising at least 90%, 95%, 96%, 97%, 98%
or 99%
identity to any one of SEQ ID NOs: 30, 31 or 32. In a further embodiment of
the
aforementioned, the antibody or antigen binding fragment thereof comprises a
heavy chain
variable region comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity to
any one of
SEQ ID NOs:26, 27, 28, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101,
102, 103, 104,
105, 106, 107, 108, 109, 110, 111, or 112; and alight chain variable region
comprising at
least 90%, 95%, 96%, 97%, 98% or 99% identity to any one of SEQ ID NOs:30, 31
or 32,
wherein any sequence variations occur in the framework regions of the
antibody. In a further
embodiment of the aforementioned, the antibody or antigen binding fragment
thereof
optionally has at least one of the following characteristics: (i) binds to
human TIGIT with a
KD value of about 1 x 10-9 M to about 1 x 1042 M as determined by surface
plasmon
-6-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or OCTET); (ii)
cross-reacts
with cynomolgous and rhesus TIGIT; (iii) blocks binding of human TIGIT to
human CD155
and human CD112; (iv) increases T cell activation; (v) stimulates antigen-
specific T-cell
production of IL-2 and IFNI', (vi) blocks induction of T cell suppression of
activation induced
by TIGIT ligation with cognate ligands CD155 and CD112.
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a heavy chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain
variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47;
(iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 4; (v)
a light chain variable region CDR2 comprising the amino acid sequence of SEQ
ID NO: 5;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 6, 57, 58, 59, 60, 61, or 62; wherein the antibody or antigen binding
fragment thereof
comprises a heavy chain variable region comprising at least 90%, 95%, 96%,
97%, 98% or
99% identity to a heavy chain variable region selected from the group
consisting of SEQ ID
NOs: 9, 10, 11, 12, 48, 49, 50, 51, 52, 53, 54, 55, and 56 and alight chain
variable region
comprising at least 90%, 95%, 96%, 97%, 98% or 99% identity to a heavy chain
variable
region selected from the group consisting of SEQ ID NOs:13, 14, 15, 16, 63,
64, 65, 66, 67,
68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, and 80. In this aforementioned
embodiment, the
sequence variations occur in the framework regions. In one embodiment, the
antibody binds
to human TIGIT with a KD value of about 1 x i0 NI to about 1 x 10-12 M as
determined by
surface plasmon resonance (e.g., BIACORE) or a similar technique (e.g. KinExa
or
OCTET).
In another embodiment, the invention provides an antibody or antigen binding
fragment that binds to human TIGIT comprising: (i) a heavy chain variable
region CDR1
comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85,
86, 87, or
86; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of SEQ ID
NO: 19; (iv) alight chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 20; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 21; and (vi) a light chain variable region CDR3 comprising the
amino acid
sequence of SEQ ID NO: 22; wherein the antibody or antigen binding fragment
thereof
comprises a heavy chain variable region comprising at least 90%, 95%, 96%,
97%, 98% or
-7-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
99% identity to a heavy chain variable region selected from the group
consisting of SEQ ID
NOs: 25, 26, 27, 28, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101,
102, 103, 104, 105,
106, 107, 108, 109, 110, 111, and 112and a light chain variable region
comprising at least
90%, 95%, 96%, 97%, 98% or 99% identity to a heavy chain variable region
selected from
the group consisting of SEQ ID NOs: 29, 30, 31, and 32. In this aforementioned
lo embodiment, the sequence variations occur in the framework regions. In
one embodiment,
the antibody binds to human TIGIT with a KD value of about 1 x 10-9M to about
1 x 10-12 M
as determined by surface plasmon resonance (e.g., BIACORE) or a similar
technique (e.g.
KinExa or OCTET).
In another embodiment, the invention also provides an antibody or antigen
binding
fragment thereof that binds to human TIGIT comprising: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii) a heavy
chain
variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45,
46, or 47;
(iv) a light chain variable region CDR1 comprising the amino acid sequence of
SEQ ID NO:
4; (v) a light chain variable region CDR2 comprising the amino acid sequence
of SEQ ID
NO: 5; and (vi) a light chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the antibody or
antigen binding
fragment thereof comprises 1, 2 or 3 amino acid substitutions in the heavy
chain CDRs (SEQ
ID NOs:1, 2, 3, 45, 46, or 47) and/or in the light chain CDRs (SEQ TD NOs: 4,
5, 6, 57, 58,
59, 60, 61, or 62). In one embodiment, the antibody binds to human TIGIT with
a KD value
of about 1 x 1019M to about 1 x 10-12 M as determined by surface plasmon
resonance (e.g.,
BIACORE) or a similar technique (e.g. KinExa or OCTET).
In another embodiment, the invention also provides an antibody or antigen
binding
fragment thereof that binds to human TIGIT comprising: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83,
84, 85, 86,
87, and 88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising the amino
acid sequence
of SEQ ID NO: 20; (v) a light chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 21; and (vi) alight chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 22. In one embodiment, the antibody or
antigen
binding fragment thereof comprises 1, 2 or 3 amino acid substitutions in the
heavy chain
CDRs (SEQ ID NOs: 17, 18, 19, 81, 82, 83, 84, 85, 86, 87, and 88) and/or in
the light chain
-8-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
CDRs (SEQ ID NOs: 20, 21, or 22). In one embodiment, the antibody binds to
human TIGIT
with a KD value of about lx 109M to about 1 x 1(112 M as determined by surface
plasmon
resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or OCTET).
In another embodiment, the invention provides an antibody or antigen binding
fragment thereof, comprising: a variable heavy chain comprising the amino acid
sequence of
SEQ ID NO: 7 and/or a variable light chain selected comprising the amino acid
sequence of
SEQ ID NO: 8, wherein the antibody or antigen binding fragment thereof binds
to human
TIGIT. In one embodiment, the antibody binds to human TIGIT with a KD value of
about 1
x 10-9M to about 1 x 102 M as determined by surface plasmon resonance (e.g.,
BIACORE)
or a similar technique (e.g. KinExa or OCTET).
In another embodiment, the invention provides an antibody or antigen binding
fragment thereof, comprising: a variable heavy chain comprising the amino acid
sequence of
SEQ ID NO: 23 and/or a variable light chain selected comprising the amino acid
sequence of
SEQ ID NO: 24, wherein the antibody or antigen binding fragment thereof binds
to human
TIGIT. In one embodiment, the antibody binds to human TIGIT with a KD value of
about 1
x 10-9 M to about 1 x 10-12 M as determined by surface plasmon resonance
(e.g., BIACORE)
or a similar technique (e.g. KinExa or OCTET),In one embodiment, the invention
relates to
an isolated antibody or antigen binding fragment that binds to human TIGIT
comprising: a
heavy chain comprising the amino acid sequence of SEQ ID NO: 7 or variant
thereof
comprising up to 30 amino acid substitutions, and/or a light chain comprising
the amino acid
sequence of SEQ ID NO: 8 comprising up to 12 amino acid substitutions.
In another embodiment, the invention relates to an isolated antibody or
antigen
binding fragment that binds to human TIGIT comprising: a heavy chain
comprising the
amino acid sequence of SEQ ID NO:10, 11, 12, 48,49, 50, 51, 52, 53, 54, 55,
56, or variant
thereof comprising up to 12 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) amino
acid substitutions,
and/or a light chain comprising the amino acid sequence of SEQ ID NO:14, 15,
16, 63, 64,
65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, or variant
thereof comprising up
to 8 (1, 2, 3, 4, 5, 6, 7 or 8) amino acid substitutions. In a further
embodiment, the invention
relates to an isolated antibody or antigen binding fragment that binds to
human TIGIT
comprising: a heavy chain comprising the amino acid sequence of SEQ ID NO: 10,
11, 12,
48, 49, 50, 51, 52, 53, 54, 55, 56, or variant thereof comprising up to 11
amino acid
substitutions, and/or a light chain comprising the amino acid sequence of SEQ
ID NO: 14, 15,
16, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80 or
variant thereof
-9-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
comprising up to 8 amino acid substitutions. In a further embodiment, the
substitutions occur
in the framework regions of the heavy and light variable chains.
In another embodiment, the invention relates to an isolated antibody or
antigen
binding fragment that binds to human TIGIT comprising: a heavy chain
comprising the
amino acid sequence of SEQ ID NO: 10 or variant thereof comprising up to 12
(1, 2, 3,4, 5,
6, 7, 8, 9, 10, 11 or 12) amino acid substitutions, and/or a light chain
comprising the amino
acid sequence of SEQ ID NO: 14 or variant thereof comprising up to 8 (1, 2, 3,
4, 5, 6, 7 or 8)
amino acid substitutions. In a further embodiment, the invention relates to an
isolated
antibody or antigen binding fragment that binds to human TIGIT comprising: a
heavy chain
comprising the amino acid sequence of SEQ ID NO: 10 or variant thereof
comprising up to
11 amino acid substitutions, and/or a light chain comprising the amino acid
sequence of SEQ
ID NO: 14 or variant thereof comprising up to 8 amino acid substitutions. In a
further
embodiment, the substitutions occur in the framework regions of the heavy and
light variable
chains.
In one embodiment, the invention relates to an isolated antibody or antigen
binding
fragment that binds to human TIGIT comprising: a heavy chain comprising the
amino acid
sequence of SEQ ID NO: 23 or variant thereof comprising up to 24 amino acid
substitutions,
and/or a light chain comprising the amino acid sequence of SEQ ID NO: 8
comprising up to
12 amino acid substitutions.
In another embodiment, the invention relates to an isolated antibody or
antigen
binding fragment that binds to human TIGIT comprising: a heavy chain
comprising the
amino acid sequence of SEQ ID NO:26, 27, 28, 89, 90, 91, 92, 93, 94, 95, 96,
97, 98, 99, 100,
101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, or variant thereof
comprising up
to 12 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) amino acid substitutions,
and/or a light chain
comprising the amino acid sequence of SEQ ID NO:30, 31, 32, or variant thereof
comprising
up to 8 (1, 2, 3, 4, 5, 6, 7 or 8) amino acid substitutions. In a further
embodiment, the
invention relates to an isolated antibody or antigen binding fragment that
binds to human
TIGIT comprising: a heavy chain comprising the amino acid sequence of SEQ ID
NO:26, 27,
28, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105,
106, 107, 108,
109, 110, 111, 112, or variant thereof comprising up to 11 amino acid
substitutions, and/or a
light chain comprising the amino acid sequence of SEQ ID NO:30, 31, 32, or
variant thereof
comprising up to 8 amino acid substitutions. In a further embodiment, the
substitutions occur
in the framework regions of the heavy and light variable chains. In another
embodiment, the
invention relates to an isolated antibody or antigen binding fragment that
binds to human
-10-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
TIGIT comprising: a heavy chain comprising the amino acid sequence of SEQ ID
NO: 26 or
variant thereof comprising up to 12 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12)
amino acid
substitutions, and/or a light chain comprising the amino acid sequence of SEQ
ID NO: 30 or
variant thereof comprising up to 8 (1, 2, 3, 4, 5, 6, 7 or 8) amino acid
substitutions. In a
further embodiment, the invention relates to an isolated antibody or antigen
binding fragment
that binds to human TIGIT comprising: a heavy chain comprising the amino acid
sequence of
SEQ ID NO: 26 or variant thereof comprising up to 11 amino acid substitutions,
and/or a
light chain comprising the amino acid sequence of SEQ ID NO: 30 or variant
thereof
comprising 4 amino acid substitutions. In a further embodiment, the
substitutions occur in
the framework regions of the heavy and variable chains.
In any of the above embodiments, the antibody or antigen binding fragment
thereof is
isolated.
In any of the above embodiments, the antibody or antigen binding fragment
thereof is
a recombinant antibody.
In any of the above embodiments, the antibody or antigen binding fragment
thereof is
a full-length antibody.
In any of the above embodiments, the antibody or antigen binding fragment
thereof is
a humanized antibody.
In any of the above embodiments, the antibody or antigen binding fragment
thereof is
a humanized antibody comprising two heavy chains and two light chains. In one
embodiment, the heavy chains are of the IgG1 isotype and the light chains are
kappa light
chains. In any of the above mentioned embodiments, the antibody or antigen
binding
fragment thereof of the invention can comprise a variable heavy region
consisting of: (a) any
of the variable heavy chains described above and (b) a leader peptide (for
example, the leader
peptide of SEQ ID NO: 40). In any of the above mentioned embodiments, the
antibody or
antigen binding fragment thereof of the invention can comprise a light heavy
region
consisting of: (a) any of the light heavy chains described above and (b) a
leader peptide (for
example, the leader peptide of SEQ ID NO: 41).
In any of the above mentioned embodiments, the antibody or antigen binding
fragment thereof of the invention is an antibody comprising any of the
variable heavy chains
described above and any human heavy chain constant domain. In one embodiment,
the
antibody or antigen binding fragment thereof of the invention is of the IgG
isotype, and
comprises a human IgGl, IgG2, IgG3 or IgG4 human heavy chain constant domain.
In one
embodiment, the antibody or antigen binding fragment thereof of the invention
comprises a
-11-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
human heavy chain IgG1 constant domain (SEQ ID NO: 44) or a variant thereof,
wherein the
variant comprises up to 20 modified amino acid substitutions. In one
embodiment, the
antibody or antigen binding fragment thereof of the invention is an antibody
comprising a
human heavy chain IgG1 constant domain comprising the amino acid sequence of
SEQ ID
NO: 44. In one embodiment, the antibody or antigen binding fragment thereof of
the
invention comprises a human heavy chain IgG1 constant domain wherein the IgG1
constant
domain is afucosylated. In one embodiment, the antibody or antigen binding
fragment
thereof of the invention comprises a human heavy chain IgG4 constant domain or
a variant
thereof, wherein the variant comprises up to 20 modified amino acid
substitutions. In another
embodiment, the antibody or antigen binding fragment thereof of the invention
comprises a
human heavy chain IgG4 constant domain, wherein the amino acid at position 228
(using EU
numbering scheme) has been substituted from Ser to Pro. In one embodiment, the
antibody
or antigen binding fragment thereof of the invention comprises a human heavy
chain IgG4
constant domain comprising the amino acid sequence of SEQ ID NO: 42.
In further embodiments, the constant domain may comprise 1, 2, 3, 4, 5, 6, 7,
8, 9, or
10 amino acid substitutions, additions, deletions, or combinations thereof as
compared to the
amino acid sequence of the native heavy chain constant domain for the human
IgGI, IgG2,
IgG3, or IgG4 isotype. In particular aspects, the constant domain may comprise
a C-terminal
lysine or may lack a C-terminal lysine.
In further embodiments, the antibody comprises an heavy chain constant domain
of a
human IgGl, IgG2, IgG3, or IgG4 isotype or variant thereof comprising 1, 2, 3,
4, 5, 6, 7, 8,
9, or 10 amino acid substitutions, additions, deletions, or combinations
thereof compared to
the amino acid sequence of the native human IgGI, IgG2, IgG3, or IgG4 isotype,
wherein the
antibody or antigen binding fragment binds TIGIT. In further aspects, the
constant domain
may comprise a C-terminal lysine or may lack a C-terminal lysine.
In further embodiments, the antibody comprises a heavy chain constant domain
of the
human IgG1 or IgG4 isotype. In a further aspect, the heavy chain constant
domain is of the
IgG4 isotype and further includes a substitution of the serine residue at
position 228 (EU
numbering) with proline, which corresponds to position 108 of SEQ ID NO:42
(Serine at
position 108). In further aspects, the constant domain may comprise a C-
terminal lysine or
may lack a C-terminal lysine.
In further embodiments, the antibody comprises a human IgG4 heavy chain
constant
domain comprising the amino acid sequence shown in SEQ ID NO:42. In further
aspects, the
Constant domain may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid
substitutions,
-12-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
additions, deletions, or combinations thereof. In further aspects, the
constant domain may
comprise a C-terminal lysine or may lack a C-terminal lysine.
In further embodiments, the antibody comprises a human IgG1 heavy chain
constant
domain comprising the amino acid sequence shown in SEQ ID NO:44. In further
aspects, the
constant domain may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid
substitutions,
additions, deletions, or combinations thereof. In further aspects, the
constant domain may
comprise a C-terminal lysine or may lack a C-terminal lysine.
In any of the above mentioned embodiments, the antibody or antigen binding
fragment thereof of the invention can comprise any of the variable light
chains described
above and human light chain constant domain. In one embodiment, the antibody
or antigen
binding fragment thereof of the invention comprises a human kappa light chain
constant
domain or a variant thereof, wherein the variant comprises up to 20 modified
amino acid
substitutions. In another embodiment, the antibody or antigen binding fragment
thereof of
the invention comprises a human lambda light chain constant domain or a
variant thereof,
wherein the variant comprises up to 20 modified amino acid substitutions. In
one
embodiment, the antibody or antigen binding fragment thereof of the invention
comprises a
human kappa light chain constant domain comprising the amino acid sequence of
SEQ ID
NO: 43.
The present invention further provides an antibody or antigen binding fragment
that
binds to an epitope on MIT comprising the amino acid sequences SSTTAQVNWEQQDQL
(SEQ ID NO:113), ICN, IYHTYPGT (SEQ ID NO:114), and GRIFL (SEQ ID NO:115). In
one embodiment, the invention provides an antibody or antigen binding fragment
that binds
to an epitope on TIGIT consisting essentially of the amino acid sequences
SSTTAQVNWEQQDQL (SEQ ID NO:113), ICN, IYHTYPGT (SEQ ID NO:114), and
GRIFL (SEQ ID NO:115).
In a further embodiment, the antibody comprises a human IgGl, IgG2, IgG3, or
IgG4
constant domain or modified derivative thereof.
In a further embodiment, the human IgGI, IgG2, IgG3, or IgG4 constant domain
is a
variant that comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid
substitutions, additions,
deletions, or combinations thereof compared to the amino acid sequence of the
native human
IgGl, IgG2, IgG3 or IgG4 isotype.
In a further embodiment, the IgGl, IgG2, IgG3, or IgG4 constant domain is a
variant
that comprises at least 1, 2, 3, or amino acid substitutions, additions,
deletions, or
-13-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
combinations thereof compared to the amino acid sequence of the native human
IgGI, IgG2,
IgG3 or Ig04 isotype.
In a further embodiment, the IgG4 constant domain is a variant that comprises
at least
a substitution of the serine at position 228 (EU numbering) or position 108 as
shown herein
with a proline residue.
1.0 In a further embodiment, the IgGl, IgG2, IgG3, or IgG4 constant domain
is a variant
that at least lacks a lysine at the C-terminus.
In a further embodiment, the antibody or antigen binding fragment comprises
variable
domain sequences comprising a framework characteristic of human antibodies.
The present invention further provides an antibody or antigen binding fragment
that
binds to an epitope on TIGIT comprising the amino acid sequences
SSTrAQVNWEQQDQL
(SEQ ID NO:113), ICN, IYHTYPGT (SEQ ID NO:114), and GRIFL (SEQ ID NO:115)
wherein the antibody or antigen binding fragment thereof comprises a human
IgG1 or IgG4
constant domain or a modified derivative thereof.
In a further embodiment, the IgG1 or IgG4 constant domain is a variant that
comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions,
additions, deletions,
or combinations thereof compared to the amino acid sequence of the native
human IgGl,
IgG2, IgG3 or IgG4 isotype.
In a further embodiment, the IgG1 or IgG4 constant domain is a variant that
comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions,
additions, deletions,
or combinations thereof compared to the amino acid sequence of the native
human IgGI,
IgG2, IgG3 or IgG4 isotype.
In a further embodiment, the IgG4 constant domain is a variant that comprises
at least
a substitution of the serine at position 228 (EU numbering) or position 108 as
shown herein
with a proline residue.
In a further embodiment, the IgG1 or IgG4 constant domain is a variant that at
least
lacks a lysine at the C-terminus.
In a further embodiment, the antibody or antigen binding fragment comprises
variable
domain sequences comprising a framework characteristic of human antibodies.
In one embodiment, the anti-TIGIT antibody of the invention comprises a
tetrameric
structure having two light chains and two heavy chains, wherein each light
chain comprises: a
variable region comprising any one of SEQ ID NOs:16, 17, 18, 63, 64, 65, 66,
67, 68, 69, 70,
71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 and a human kappa light chain
constant region (SEQ
ID NO:43); and each heavy chain comprises: a variable region comprising any
one of SEQ
-14-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
ID NOs:9, 10, 11, 12, 48,49, 50, 51, 52, 53, 54, 55, or 56, and a human IgG1
constant region
(SEQ ID NO: 44).
In one embodiment, the anti-TIGIT antibody of the invention comprises a
tetrameric
structure having two light chains and two heavy chains, wherein each light
chain comprises: a
variable region comprising any one of SEQ ID NOs:29, 30, 31, or 32 and a human
kappa
light chain constant region (SEQ ID NO:43); and each heavy chain comprises: a
variable
region comprising any one of SEQ ID NOs:25, 26, 27, 28, 89, 90, 91, 92, 93,
94, 95, 96, 97,
98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, or 112,
and a human IgG1
constant region (SEQ ID NO: 44).
In one embodiment, the anti-TIGIT antibody of the invention comprises a
tetrameric
structure having two light chains and two heavy chains, wherein each light
chain comprises: a
variable region comprising any one of SEQ ID NOs:13-16 or 29-32 and a human
kappa light
chain constant region (SEQ ID NO:43): and each heavy chain comprises: a
variable region
comprising any one of SEQ ID NOs:9-13 or 25-28, a human IgG1 constant region
(SEQ ID
NO: 44).
In any of the above mentioned embodiments, the anti-TIGTT antibody or antigen
binding fragment thereof of the invention can be conjugated to at least one
therapeutic agent.
In one embodiment, the wherein the therapeutic agent comprises a second
antibody or
fragment thereof, an immunomodulator, a hormone, a cytotoxic agent, an enzyme,
a
radionuclide, a second antibody conjugated to at least one immunomodulator,
enzyme,
radioactive label, hormone, antisense oligonucleotide, or cytotoxic agent, or
a combination
thereof.
The invention also provides isolated polypeptides comprising the amino acid
sequence of any one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,
14, 15, 16, 17, 18,
19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 45, 46, 47, 48, 49,
50, 51, 52, 53, 54, 55,
56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74,
75, 76, 77, 78, 79, 80,
81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99,
100, 101, 102, 103,
104, 105, 106, 107, 108, 109, 110, 111, 112, or a fragment of any said
sequences.
The invention also provides isolated nucleic acids encoding anyone of the anti-
TIGIT
antibodies or antigen binding fragments of the invention. In one embodiment,
the invention
provides isolated nucleic acids encoding anyone of the polypeptides of SEQ ID
NOs: 1, 2, 3,
4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,
25, 26, 27, 28, 29, 30,
31, 32, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61,
62, 63, 64, 65, 66, 67,
68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86,
87, 88, 89, 90, 91, 92,
-15-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109,
110, 111, or 112,
wherein said polypeptides can optionally comprise a leader sequence. The
invention also
provides expression vectors comprising a nucleic acid encoding anyone of the
polypeptides
of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18,
19, 20, 21, 22,23,
24, 25, 26, 27, 28, 29, 30, 31, 32, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54,
55, 56, 57, 58, 59, 60,
61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79,
80, 81, 82, 83, 84, 85,
86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103,
104, 105, 106, 107,
108, 109, 110, 111, or 112 (wherein said polypeptides can optionally comprise
a leader
sequence). These isolated nucleic acids and the expression vectors comprising
them may be
used to express the antibodies of the invention or antigen binding fragments
thereof in
recombinant host cells. Thus, the invention also provides host cells
comprising isolated
nucleic acids encoding anyone of the polypeptides of SEQ ID NOs: 1, 2, 3, 4,
5, 6, 7, 8, 9, 10,
11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29,
30, 31, 32, 45, 46, 47,
48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66,
67, 68, 69, 70, 71, 72,
73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91,
92, 93, 94, 95, 96, 97,
98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 1 1 1 , or 112,
(wherein said
polypeptides can optionally comprise a leader sequence). In one embodiment,
the host cell is
Chinese hamster oval), cell. In one embodiment, the host cell is a yeast cell,
for example a
Pichia cell or a Pichia pastoris host cell.
The invention also provides pharmaceutical compositions comprising an antibody
or
antigen binding fragment of the invention and a pharmaceutically acceptable
carrier or
diluent. In one embodiment, the composition comprises a further therapeutic
agent. In one
embodiment, the further therapeutic agent is selected from the group
consisting of: an anti-
PDI antibody or an antigen binding fragment thereof; an anti-LAG3 antibody or
an antigen
binding fragment thereof; an anti-VISTA antibody or an antigen binding
fragment thereof;
an anti-BTLA antibody or an antigen binding fragment thereof; an anti-TIM3
antibody or an
antigen binding fragment thereof; an anti-CTLA4 antibody or an antigen binding
fragment
thereof; an anti-HVEM antibody or an antigen binding fragment thereof; an anti-
CD27
antibody or an antigen binding fragment thereof; an anti-CD137 antibody or an
antigen
binding fragment thereof; an anti-0X40 antibody or an antigen binding fragment
thereof; an
anti-CD28 antibody or an antigen binding fragment thereof; an anti-PDL1
antibody or an
antigen binding fragment thereof; an anti-PDL2 antibody or an antigen binding
fragment
thereof; an anti-GITR antibody or an antigen binding fragment thereof; an anti
-ICOS
antibody or an antigen binding fragment thereof; an anti-STRPa antibody or an
antigen
-16-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
binding fragment thereof; an anti-ILT2 antibody or antigen binding fragment
thereof; an
anti-ILT3 antibody or antigen binding fragment thereof; an anti-ILT4 antibody
or antigen
binding fragment thereof; and an anti-ILT5 antibody or an antigen binding
fragment thereof;
an anti 4-1BB antibody or an antigen binding fragment thereof. In one
embodiment, the anti-
PD1 antibody or an antigen binding fragment thereof; is selected from the
group consisting
of: pembrolizumab or an antigen binding fragment thereof and nivolumab or an
antigen
binding fragment thereof.
In one embodiment, the anti-TIGIT antibody or antigen binding fragment of the
invention comprises: (i) a heavy chain variable region CDR1 comprising the
amino acid
sequence of SEQ ID NO: 1; (ii) a heavy chain variable region CDR2 comprising
the amino
acid sequence of SEQ ID NO: 2; (iii) a heavy chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 3, 45, 46, or 47; (iv) a light chain
variable region CDR1
comprising the amino acid sequence of SEQ ID NO: 4; (v) a light chain variable
region
CDR2 comprising the amino acid sequence of SEQ ID NO: 5; and (vi) a light
chain variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 6, 57, 58, 59,
60, 61, or
62. In one embodiment, the anti-TIGIT antibody or antigen binding fragment
thereof
comprises: (i) a heavy chain variable region CDR1 comprising the amino acid
sequence of
SEQ ID NO: 17; (ii) a heavy chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85, 86, 87, or 88; (iii) a heavy
chain variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 19; (iv) alight
chain
variable region CDRI comprising the amino acid sequence of SEQ ID NO: 20; (v)
a light
chain variable region CDR2 comprising the amino acid sequence of SEQ ID NO:
21; and (vi)
a light chain variable region CDR3 comprising the amino acid sequence of SEQ
ID NO: 22.
In one embodiment, the invention provides a composition comprising: (i) an
anti-
TIGIT antibody or antigen binding fragment of the invention; and (ii) an anti-
PD1 antibody
comprising the heavy chain sequence of SEQ ID NO: 36 and the light chain
variable
sequence of SEQ ID NO: 37. In another embodiment, the invention provides a
composition
comprising: (a) an anti-TIGIT antibody or antigen binding fragment of the
invention; and (b)
an anti-PD1 antibody comprising the heavy chain sequence of SEQ ID NO: 38 and
the light
chain variable sequence of SEQ ID NO: 39. In one embodiment, the anti-PD1
antibody is
administered prior to the administration of an anti-TIGIT antibody. In one
embodiment, the
anti-PD1 antibody is administered 4-10 days prior to the administration of the
anti-TIGIT
antibody. In one embodiment, pretreatment treatment with anti-PD1 antibody may
modulate
immune cells resulting in enhanced Fc-mediated function of the anti-MIT
antibodies. In
-17-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
one embodiment, the anti-TIGIT antibody or antigen binding fragment of the
invention
comprises: (i) a heavy chain variable region CDR1 comprising the amino acid
sequence of
SEQ ID NO: 1; (ii) a heavy chain variable region CDR2 comprising the amino
acid sequence
of SEQ ID NO: 2; (iii) a heavy chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 3, 45, 46, or 47; (iv) alight chain variable region
CDR1 comprising
the amino acid sequence of SEQ ID NO: 4; (v) a light chain variable region
CDR2
comprising the amino acid sequence of SEQ ID NO: 5; and (vi) a light chain
variable region
CDR3 comprising the amino acid sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61,
or 62. In
one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof
comprises: (i)
a heavy chain variable region CDR1 comprising the amino acid sequence of SEQ
ID NO: 17;
(ii) a heavy chain variable region CDR2 comprising the amino acid sequence of
SEQ ID NO:
18, 81, 82, 83, 84, 85, 86, 87, or 88; (iii) a heavy chain variable region
CDR3 comprising the
amino acid sequence of SEQ ID NO: 19; (iv) a light chain variable region CDR1
comprising
the amino acid sequence of SEQ ID NO: 20; (v) a light chain variable region
CDR2
comprising the amino acid sequence of SEQ ID NO: 21; and (vi) a light chain
variable region
CDR3 comprising the amino acid sequence of SEQ ID NO: 22.
The invention also comprises a combination comprising an anti-TIGIT antibody
or
antigen binding fragment of the invention, in combination with one, two or
more therapeutic
agents; wherein the second therapeutic agent is selected from the group
consisting of: an anti-
PDI antibody or an antigen binding fragment thereof; an anti-LAG3 antibody or
an antigen
binding fragment thereof; an anti-VISTA antibody or an antigen binding
fragment thereat
an anti-BTLA antibody or an antigen binding fragment thereof; an anti-TIM3
antibody or an
antigen binding fragment thereof; an anti-CTLA4 antibody or an antigen binding
fragment
thereof; an anti-HVEM antibody or an antigen binding fragment thereof; an anti-
CD27
antibody or an antigen binding fragment thereof; an anti-CD137 antibody or an
antigen
binding fragment thereof; an anti-OX40 antibody or an antigen binding fragment
thereof; an
anti-CD28 antibody or an antigen binding fragment thereof; an anti-PDL I
antibody or an
antigen binding fragment thereof; an anti-PDL2 antibody or an antigen binding
fragment
thereof; an anti-GITR antibody or an antigen binding fragment thereof; an anti-
ICOS
antibody or an antigen binding fragment thereof; an anti-SIRPa antibody or an
antigen
binding fragment thereof; an anti-ILT2 antibody or antigen binding fragment
thereof; an anti-
ILT3 antibody or antigen binding fragment thereof; an anti-ILT4 antibody or
antigen binding
fragment thereof; an anti-ILT5 antibody or an antigen binding fragment
thereof; and an anti
4-1BB antibody or an antigen binding fragment thereof. In one embodiment, the
anti-'TIGIT
-18-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
antibody or antigen binding fragment of the invention comprises: (i) a heavy
chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy
chain
variable region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii)
a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3,
45, 46,
or 47; (iv) a light chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 4; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 5; and (vi) a light chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the
anti-TIGIT
antibody or antigen binding fragment thereof comprises: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83,
84, 85, 86,
87, or 88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising the amino
acid sequence
of SEQ ID NO: 20; (v) a light chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 21; and (vi) a light chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 22.
The invention also provides a vessel or injection device comprising anyone of
the
anti-TIGIT antibodies or antigen binding fragments of the invention. In one
embodiment, the
anti-TIGIT antibody or antigen binding fragment of the invention comprises:
(i) a heavy
chain variable region CDR1 comprising the amino acid sequence of SEQ ID NO: 1;
(ii) a
heavy chain variable region CDR2 comprising the amino acid sequence of SEQ ID
NO: 2;
(iii) a heavy chain variable region CDR3 comprising the amino acid sequence of
SEQ ID
NO: 3, 45, 46, or 47; (iv) a light chain variable region CDR1 comprising the
amino acid
sequence of SEQ ID NO: 4; (v) a light chain variable region CDR2 comprising
the amino
acid sequence of SEQ ID NO: 5; and (vi) a light chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one
embodiment, the
anti-TIGIT antibody or antigen binding fragment thereof comprises: (i) a heavy
chain
variable region CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii)
a heavy
chain variable region CDR2 comprising the amino acid sequence of SEQ ID NO:
18, 81, 82,
83, 84, 85, 86, 87, or 88; (iii) a heavy chain variable region CDR3 comprising
the amino acid
sequence of SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising
the amino
acid sequence of SEQ ID NO: 20; (v) a light chain variable region CDR2
comprising the
amino acid sequence of SEQ ID NO: 21; and (vi) a light chain variable region
CDR3
comprising the amino acid sequence of SEQ ID NO: 22.
-19-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
The invention also provides a method of producing an anti-TIGIT antibody or
antigen
binding fragment of the invention comprising: culturing a host cell comprising
a
polynucleotide encoding a heavy chain and/or light chain of an antibody of the
invention (or
an antigen binding fragment thereof) under conditions favorable to expression
of the
polynucleotide; and optionally, recovering the antibody or antigen binding
fragment from the
host cell and/or culture medium. In one embodiment, the polynucleotide
encoding the heavy
chain and the polynucleotide encoding the light chain are in a single vector.
In another
embodiment, the polynucleotide encoding the heavy chain and the polynucleotide
encoding
the light chain are in different vectors. In one embodiment, the
polynucleotide encoding the
heavy chain and the polynucleotide encoding the light chain encode an antibody
or antigen
binding fragment comprising: (i) a heavy chain variable region CDR1 comprising
the amino
acid sequence of SEQ ID NO: 1; (ii) a heavy chain variable region CDR2
comprising the
amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain variable region CDR3
comprising
the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47; (iv) a light chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO:4; (v) a light chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO:5; and (vi) a light chain
variable
region CDR3 comprising the amino acid sequence of SEQ ID NO:6, 57, 58, 59, 60,
61, or 62.
In another embodiment, the polynucleotide encoding the heavy chain and the
polynucleotide
encoding the light chain encode an antibody or antigen binding fragment
comprising: (i) a
heavy chain variable region CDR] comprising the amino acid sequence of SEQ ID
NO:17;
(ii) a heavy chain variable region CDR2 comprising the amino acid sequence of
SEQ ID NO:
18, 81, 82, 83, 84, 85, 86, 87, or 88; (iii) a heavy chain variable region
CDR3 comprising the
amino acid sequence of SEQ ID NO: 19; (iv) alight chain variable region CDR1
comprising
the amino acid sequence of SEQ ID NO: 20; (v) a light chain variable region
CDR2
comprising the amino acid sequence of SEQ ID NO: 21; and (vi) a light chain
variable region
CDR3 comprising the amino acid sequence of SEQ ID NO: 22.
The invention also provides a method of treating cancer in a subject in need
thereof,
comprising administering to the subject an effective amount of an anti-TIGIT
antibody or
antigen binding fragment of the invention, optionally in association with a
further therapeutic
agent or therapeutic procedure. In one embodiment, the subject been treated is
a human
subject. In one embodiment, the further therapeutic agent is selected from the
group
consisting of: an anti-PD1 antibody or an antigen binding fragment thereof; an
anti-LAG3
antibody or an antigen binding fragment thereof; an anti-VISTA antibody or an
antigen
binding fragment thereof, an anti -BTLA antibody or an antigen binding
fragment thereof; an
-20-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
anti-TIM3 antibody or an antigen binding fragment thereof; an anti-CTLA4
antibody or an
antigen binding fragment thereof; an anti-HVEM antibody or an antigen binding
fragment
thereof; an anti-CD27 antibody or an antigen binding fragment thereof; an anti-
CD137
antibody or an antigen binding fragment thereof; an anti-0X40 antibody or an
antigen
binding fragment thereof; an anti-CD28 antibody or an antigen binding fragment
thereof; an
anti-PDL1 antibody or an antigen binding fragment thereof; an anti-PDL2
antibody or an
antigen binding fragment thereof; an anti-GITR antibody or an antigen binding
fragment
thereof; an anti-ICOS antibody or an antigen binding fragment thereof; an anti-
SIRPa
antibody or an antigen binding fragment thereof an anti-ILT2 antibody or
antigen binding
fragment thereof; an anti-ILT3 antibody or antigen binding fragment thereof;
an anti-ILT4
antibody or antigen binding fragment thereof; an anti-ILT5 antibody or an
antigen binding
fragment thereof; and an anti-4-1BB antibody or an antigen binding fragment
thereof. In one
embodiment, the anti-PD1 antibody or an antigen binding fragment thereof is
selected from
the group consisting of: pembroliztunab or an antigen binding fragment thereof
and
nivoluinab or an antigen binding fragment thereof In one embodiment, the anti-
TIGIT
antibody or antigen binding fragment of the invention comprises: (i) a heavy
chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy
chain
variable region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii)
a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3,
45, 46,
or 47; (iv) a light chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 4; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 5; and (vi) a light chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the
anti-MIT
antibody or antigen binding fragment thereof comprises: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83,
84, 85, 86,
87, or 88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising the amino
acid sequence
of SEQ ID NO: 20; (v) a light chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 21; and (vi) a light chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 22.
The invention also provides a method of treating cancer in a subject in need
thereof,
comprising administering to the subject an effective amount of an anti-TIGIT
antibody or
antigen binding fragment of the invention, and further administering an anti-
PD1 antibody or
-21-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
an antigen binding fragment thereof. In one embodiment, the anti-PD1 antibody
or an
antigen binding fragment thereof is selected from the group consisting of:
pembrolizumab or
an antigen binding fragment thereof and nivolumab or an antigen binding
fragment thereof.
In one embodiment, the anti-TIGIT antibody or antigen binding fragment of the
invention
comprises: (i) a heavy chain variable region CDR1 comprising the amino acid
sequence of
SEQ ID NO: 1; (ii) a heavy chain variable region CDR2 comprising the amino
acid sequence
of SEQ ID NO: 2; (iii) a heavy chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 3, 45, 46, or 47; (iv) a light chain variable region
CDR1 comprising
the amino acid sequence of SEQ ID NO: 4; (v) a light chain variable region
CDR2
comprising the amino acid sequence of SEQ ID NO: 5; and (vi) a light chain
variable region
CDR3 comprising the amino acid sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61,
or 62. In
one embodiment, the anti-TIGIT antibody or antigen binding fragment thereof
comprises: (i)
a heavy chain variable region CDR1 comprising the amino acid sequence of SEQ
ID NO: 17;
(ii) a heavy chain variable region CDR2 comprising the amino acid sequence of
SEQ ID NO:
18, 81, 82, 83, 84, 85, 86, 87, or 88; (iii) a heavy chain variable region
CDR3 comprising the
amino acid sequence of SEQ ID NO: 19; (iv) alight chain variable region CDR1
comprising
the amino acid sequence of SEQ ID NO: 20; (v) a light chain variable region
CDR2
comprising the amino acid sequence of SEQ ID NO: 21; and (vi) a light chain
variable region
CDR3 comprising the amino acid sequence of SEQ ID NO: 22.
The invention also provides a method of treating an infection or infectious
disease in a
subject, comprising administering to the subject an effective amount of an
antibody or
antigen binding fragment of the invention, optionally in association with a
further therapeutic
agent or therapeutic procedure. In one embodiment, the subject been treated is
a human
subject. In one embodiment, the further therapeutic agent is selected from the
group
consisting of: an anti-PD1 antibody or an antigen binding fragment thereof; an
anti-LAG3
antibody or an antigen binding fragment thereof; an anti-VISTA antibody or an
antigen
binding fragment thereof; an anti -BTLA antibody or an antigen binding
fragment thereof; an
anti-TIM3 antibody or an antigen binding fragment thereof; an anti-CTLA4
antibody or an
antigen binding fragment thereof; an anti-HVEM antibody or an antigen binding
fragment
thereof; an anti-CD27 antibody or an antigen binding fragment thereof; an anti-
CD137
antibody or an antigen binding fragment thereof; an anti-0X40 antibody or an
antigen
binding fragment thereof; an anti-CD28 antibody or an antigen binding fragment
thereof; an
anti-PDL1 antibody or an antigen binding fragment thereof; an anti-PDL2
antibody or an
antigen binding fragment thereof; an anti-GITR antibody or an antigen binding
fragment
-22-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
thereof; an anti-ICOS antibody or an antigen binding fragment thereof; an anti-
SIRPa
antibody or an antigen binding fragment thereof; an anti-ILT2 antibody or
antigen binding
fragment thereof; an anti-ILT3 antibody or antigen binding fragment thereof;
an anti-ILT4
antibody or antigen binding fragment thereof; an anti-ILT5 antibody or an
antigen binding
fragment thereof; and an anti-4-1BB antibody or an antigen binding fragment
thereof. In one
embodiment, the anti-PD I antibody or an antigen binding fragment thereof; is
selected from
the group consisting of: pembroliannab or an antigen binding fragment thereof
and
nivolinnab or an antigen binding fragment thereof In one embodiment, the anti-
TIGIT
antibody or antigen binding fragment of the invention comprises: (i) a heavy
chain variable
region CDRI comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy
chain
variable region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii)
a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3,
45, 46,
or 47; (iv) a light chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 4; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 5; and (vi) a light chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the
anti-MIT
antibody or antigen binding fragment thereof comprises: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83,
84, 85, 86,
87, or 88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising the amino
acid sequence
of SEQ ID NO: 20; (v) a light chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 21; and (vi) alight chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 22.
The invention also provides a vaccine comprising an antibody or antigen
binding
fragment of the invention. In one embodiment, the anti-TIGIT antibody or
antigen binding
fragment of the invention comprises: (i) a heavy chain variable region CDR1
comprising the
amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain variable region CDR2
comprising
the amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain variable region
CDR3
comprising the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47; (iv) a
light chain
variable region CDR1 comprising the amino acid sequence of SEQ ID NO: 4; (v) a
light
chain variable region CDR2 comprising the amino acid sequence of SEQ ID NO: 5;
and (vi)
a light chain variable region CDR3 comprising the amino acid sequence of SEQ
ID NO: 6,
57, 58, 59, 60, 61, or 62. In one embodiment, the anti-TIGIT antibody or
antigen binding
-23-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
fragment thereof comprises: (i) a heavy chain variable region CDR1 comprising
the amino
acid sequence of SEQ ID NO: 17; (ii) a heavy chain variable region CDR2
comprising the
amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85, 86, 87, or 88; (iii)
a heavy chain
variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 19; (iv)
alight
chain variable region CDR1 comprising the amino acid sequence of SEQ ID NO:
20; (v) a
light chain variable region CDR2 comprising the amino acid sequence of SEQ ID
NO: 21;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 22. In one embodiment, the vaccine further comprises an antigen.
The invention also provides a method for detecting the presence of a TIGIT
peptide or
a fragment thereof in a sample comprising contacting the sample with an
antibody or antigen
binding fragment thereof of the invention and detecting the presence of a
complex between
the antibody or fragment and the peptide; wherein detection of the complex
indicates the
presence of the TIGIT peptide. In one embodiment, the anti-'TIGIT antibody or
antigen
binding fragment of the invention comprises: (i) a heavy chain variable region
CDR1
comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain
variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47;
(iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 4; (v)
a light chain variable region CDR2 comprising the amino acid sequence of SEQ
ID NO: 5;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the anti-TIGIT antibody
or antigen
binding fragment thereof comprises: (i) a heavy chain variable region CDR1
comprising the
amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain variable region CDR2
comprising
the amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85, 86, 87, or 88;
(iii) a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO:
19; (iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 20; (v)
alight chain variable region CDR2 comprising the amino acid sequence of SEQ ID
NO: 21;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 22.
The invention also provides a method of increasing the activity of an immune
cell,
comprising contacting the immune cell with any one of the antibodies or
antigen binding
fragments of the invention. In one embodiment, the invention provides a method
of
increasing the activity of an immune cell, comprising administering to a
subject in need
thereof an effective amount of an antibody or antigen binding fragments of the
invention. In
-24-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
one embodiment, the method is used for: the treatment of cancer, the treatment
of an
infection or infectious disease, or as a vaccine adjuvant. In one embodiment,
the increase in
activity of an immune cell can be detected by measuring the proliferation of
the immune cell.
For example, an increase in activity of a T cell can be detected by measuring
the proliferation
of the T cell. In one embodiment, the increase in activity of an immune cell
can be detected
by measuring T cell activation ex vivo in a sample derived from the subject.
In one
embodiment, the increase in T cell activity is determined by: (i) measuring
mixed
lymphocyte reactions or direct anti-CD3 mAb stimulation of T cell receptor
(TCR) signaling
to induce production of a cytokine selected from the group consisting of: 1L-
2, 'TNFa, IL-17,
IFNy, IL-113, GM-CSF, RANTES, IL-6, IL-8, IL-5 and IL-13; (ii) measuring SEB
induced
production of one or more cytokines selected from the group consisting of: IL-
2, TNFix, IL-
17, IFNy, GM-CSF, RANTES, IL-6, IL-8, IL-5 and IL-13; or (iii) measuring TT
induced
production of a cytokine selected from the group consisting of: IL-2, TNFa, IL-
17, IFNy,
GM-CSF, RANTES, 1L-6, IL-8, IL-5 and IL-13. In one embodiment, the anti-TIGIT
antibody or antigen binding fragment of the invention comprises: (i) a heavy
chain variable
region CDR1 comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy
chain
variable region CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii)
a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO: 3,
45, 46,
or 47; (iv) a light chain variable region CDR1 comprising the amino acid
sequence of SEQ
ID NO: 4; (v) a light chain variable region CDR2 comprising the amino acid
sequence of
SEQ ID NO: 5; and (vi) a light chain variable region CDR3 comprising the amino
acid
sequence of SEQ ID NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the
anti-TIGIT
antibody or antigen binding fragment thereof comprises: (i) a heavy chain
variable region
CDR1 comprising the amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain
variable
region CDR2 comprising the amino acid sequence of SEQ ID NO: 18, 81, 82, 83,
84, 85, 86,
87, or 88; (iii) a heavy chain variable region CDR3 comprising the amino acid
sequence of
SEQ ID NO: 19; (iv) a light chain variable region CDR1 comprising the amino
acid sequence
of SEQ ID NO: 20; (v) a light chain variable region CDR2 comprising the amino
acid
sequence of SEQ ID NO: 21; and (vi) a light chain variable region CDR3
comprising the
amino acid sequence of SEQ ID NO: 22.
The invention also comprises a method of treating cancer or infectious disease
in a
subject, comprising administering to the subject an effective amount of an
antagonist anti-
TIGIT antibody of the invention and an antagonist anti-PD1 antibody, wherein
the anti-
TIGIT antibody is afucosylated. In one embodiment, the anti-TTGIT antibody or
antigen
-25-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
binding fragment of the invention comprises: (i) a heavy chain variable region
CDR1
comprising the amino acid sequence of SEQ ID NO: 1; (ii) a heavy chain
variable region
CDR2 comprising the amino acid sequence of SEQ ID NO: 2; (iii) a heavy chain
variable
region CDR3 comprising the amino acid sequence of SEQ ID NO: 3, 45, 46, or 47;
(iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 4; (v)
a light chain variable region CDR2 comprising the amino acid sequence of SEQ
ID NO: 5;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 6, 57, 58, 59, 60, 61, or 62. In one embodiment, the anti-TIGIT antibody
or antigen
binding fragment thereof comprises: (i) a heavy chain variable region CDRI
comprising the
amino acid sequence of SEQ ID NO: 17; (ii) a heavy chain variable region CDR2
comprising
the amino acid sequence of SEQ ID NO: 18, 81, 82, 83, 84, 85, 86, 87, or 88;
(iii) a heavy
chain variable region CDR3 comprising the amino acid sequence of SEQ ID NO:
19; (iv) a
light chain variable region CDR1 comprising the amino acid sequence of SEQ ID
NO: 20; (v)
a light chain variable region CDR2 comprising the amino acid sequence of SEQ
ID NO: 21;
and (vi) a light chain variable region CDR3 comprising the amino acid sequence
of SEQ ID
NO: 22.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows binding of antibodies of the invention to human and cynolrhesus
TIGIT.
Figure 2 shows that antibodies of the invention block hCD155 interaction with
hTIGIT as determined by a cell-based ELISA blocking assay.
Figure 3 shows the activity of antibodies of the invention in an in vitro T
cell assay.
Figure 4 shows a deuterium labeling difference heatmap of the human TIGIT
amino
acid residues bound by the mouse anti-human TIGIT 14D7 antibody.
DETAILED DESCRIPTION
Abbreviations
Throughout the detailed description and examples of the invention the
following
abbreviations will be used:
ADCC Antibody-dependent cellular cytotoxicity
CDC Complement-dependent cytotoxicity
-26-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
CDR Complementarity determining region in the immunoglobulin variable
regions,
defined using the Kabat numbering system
CHO Chinese hamster ovaiy
ELISA Enzyme-linked immunosorbant assay
FR Antibody framework region: the immunoglobulin variable regions
excluding
the CDR regions.
HRP Horseradish peroxidase
IFN interferon
IC50 concentration resulting in 50% inhibition
IgG Immunoglobulin G
Kabat An immunoglobulin alignment and numbering system pioneered by Elvin
A.
Kabat ((1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public
Health Service, National Institutes of Health, Bethesda, Md.)
inAb or Mab or MAb Monoclonal antibody
SEB Staphylococcus Enterotoxin B
Tr Tetanus toxoid
V region The segment of IgG chains which is variable in sequence between
different
antibodies. It extends to Kabat residue 109 in the light chain and 113 in the
heavy chain.
VH Immunoglobulin heavy chain variable region
VK Immunoglobulin kappa light chain variable region
Definitions
So that the invention may be more readily understood, certain technical and
scientific
terms are specifically defined below. Unless specifically defined elsewhere in
this document,
all other technical and scientific terms used herein have the meaning commonly
understood
by one of ordinary skill in the art to which this invention belongs.
As used herein, including the appended claims, the singular forms of words
such as
"a," "an," and "the," include their corresponding plural references unless the
context clearly
dictates otherwise.
"Administration" and "treatment," as it applies to an animal, human,
experimental
subject, cell, tissue, organ, or biological fluid, refers to contact of an
exogenous
pharmaceutical, therapeutic, diagnostic agent, or composition to the animal,
human, subject,
cell, tissue, organ, or biological fluid. Treatment of a cell encompasses
contact of a reagent
-27-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
to the cell, as well as contact of a reagent to a fluid, where the fluid is in
contact with the cell.
"Administration" and "treatment" also means in vitro and ex vivo treatments,
e.g., of a cell, by
a reagent, diagnostic, binding compound, or by another cell.
"Treat" or "treating" means to administer a therapeutic agent, such as a
composition
containing any of the antibodies or antigen-binding fragments of the present
invention,
internally or externally to a subject or patient having one or more disease
symptoms, or being
suspected of having a disease, for which the agent has therapeutic activity.
Typically, the
agent is administered in an amount effective to alleviate one or more disease
symptoms in the
treated subject or population, whether by inducing the regression of or
inhibiting the
progression of such symptom(s) by any clinically measurable degree. The amount
of a
therapeutic agent that is effective to alleviate any particular disease
symptom may vary
according to factors such as the disease state, age, and weight of the
patient, and the ability of
the drug to elicit a desired response in the subject. Whether a disease
symptom has been
alleviated can be assessed by any clinical measurement typically used by
physicians or other
skilled healthcare providers to assess the severity or progression status of
that symptom.
TIGIT
The term TIGIT includes human TIGIT, cynomolgous monkey TIGIT and rhesus
TIGIT as well as fragments thereof such as the mature fragment thereof lacking
the signal
peptide. In an embodiment of the invention, the amino acid sequence of human
TIGIT
comprises the amino acid sequence disclosed in amino acid residues 25-244 of
Genbank
Accession Number NP 776160.2 (SEQ ID NO: 33). (Amino acid residues 1-24 of SEQ
ID
NO:33 correspond to a leader peptide that is removed from the mature 'MIT
protein.)
In an embodiment of the invention, the amino acid sequence of cynomolgous
monkey,
e.g., Macaea.fascieularis TIGIT comprises the amino acid sequence disclosed in
(SEQ ID
NO: 34); see also Genbank Accession no. XP 005548157. The amino acid sequence
of
rhesus monkey TIGIT is identical to the amino acid sequence of cynomolgous
monkey
TIGIT. (Amino acid residues 1-24 of SEQ ID NO:34 correspond to a leader
peptide that is
removed from the mature TIGIT protein.)
Anti-TIGIT Antibodies and Antigen-Binding Fragments Thereof
The present invention provides antibodies or antigen-binding fragments thereof
that
bind human TIGIT and uses of such antibodies or fragments. In some
embodiments, the anti-
TIGIT antibodies are isolated.
-28-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
As used herein, an anti-TIGIT antibody or antigen-binding fragment thereof
refers to
an antibody or antigen-binding fragment thereof that specifically binds to
human TIGIT. An
antibody or antigen-binding fragment thereof that "specifically binds to human
TIGIT" is an
antibody or antigen-binding fragment thereof that binds to human TIGIT with a
KD of about
1 nM or a higher affinity (e.g., 1 nM-2 pM, 1 nM, 100 pM, 10 pM or 2 pM). For
example, an
antibody that "specifically binds" human TIGIT does not bind to human CD226,
human
CD155 and human CD112. As a further example, an antibody or antigen-binding
fragment
that specifically binds to human TIGIT may bind to a FLAGS-tagged form of
human TIGIT
but will not bind to other FLAGS-tagged proteins that lack human TIGIT
epitopes. In one
embodiment, the antibody of the invention which specifically binds to human
TIGIT is also
cross-reactive with cynomolgus and rhesus TIGIT. As used herein "cross-
reactivity" refers
to the ability of an antibody to react with a homologous protein from other
species, which can
be determine using any assay known in the art. Whether an antibody
specifically binds to
human TIGIT can be determined using any assay known in the art. Examples of
assays
known in the art to determining binding affinity and cross-reactivity include
surface plasmon
resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or OCTET).
The present invention includes anti-TIGIT antibodies and methods of use
thereof. As
used herein, the term "antibody" refers to any form of antibody that exhibits
the desired
biological activity. Thus, it is used in the broadest sense and specifically
covers, but is not
limited to, monoclonal antibodies (including full length monoclonal antibodies
comprising
two light chains and two heavy chains), polyclonal antibodies, multispecific
antibodies (e.g.,
bispecific antibodies), humanized antibodies, fully human antibodies and
chimeric antibodies.
The present invention includes non-human parental (e.g. mouse and rodent) anti-

TIGIT antibodies and antigen-binding fragments thereof and methods of use
thereof. These
antibodies may be modified for an intended use, such as humanization of an
antibody for use
as a human therapeutic antibody or fragment.
The present invention includes anti-TIGIT antigen-binding fragments and
methods of
use thereof. As used herein, unless otherwise indicated, "antibody fragment"
or "antigen-
binding fragment" refers to antigen-binding fragments of antibodies, i.e.
antibody fragments
that retain the ability to bind specifically to the antigen bound by the full-
length antibody, e.g.
fragments that retain one or more CDR regions. Examples of antigen-binding
fragments
include, but are not limited to, Fab, Fab', F(ab)2, and Fv fragments;
diabodies; linear
antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and
multispecific
antibodies formed from full-length antibody chains or from antibody fragments.
-29-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
The present invention includes anti-TIGIT Fab fragments and methods of use
thereof.
A "Fab fragment" is comprised of one light chain and the C111 and variable
regions of one
heavy chain. The heavy chain of a Fab molecule cannot form a disulfide bond
with another
heavy chain molecule. An "Fab fragment" can be the product of papain cleavage
of an
antibody.
The present invention includes anti- TIGIT antibodies and antigen-binding
fragments
thereof which comprise an Fc region and methods of use thereof. An "Fe" region
contains
two heavy chain fragments comprising the CHI and CH2 domains of an antibody.
The two
heavy chain fragments are held together by two or more disulfide bonds and by
hydrophobic
interactions of the CH3 domains.
The present invention includes anti-TIG1T Fab fragments and methods of use
thereof.
A "Fab fragment" contains one light chain and a portion or fragment of one
heavy chain that
contains the VH domain and the C H1 domain and also the region between the
C111 and C H2
domains, such that an interchain disulfide bond can be formed between the two
heavy chains
of two Fab fragments to form a F(ab') 2 molecule.
The present invention includes anti-TIGIT F(ab')2 fragments and methods of use
thereof. A "F(alTh fragment" contains two light chains and two heavy chains
containing a
portion of the constant region between the CHI and CH2 domains, such that an
interchain
disulfide bond is formed between the two heavy chains. A F(ab') 2fragment thus
is
composed of two Fab' fragments that are held together by a disulfide bond
between the two
heavy chains. An "F(a1:02 fragment" can be the product of pepsin cleavage of
an antibody.
The present invention includes anti-TIGIT Fv fragments and methods of use
thereof.
The "Fv region" comprises the variable regions from both the heavy and light
chains, but
lacks the constant regions.
The present invention includes anti-TIG1T seFv fragments and methods of use
thereof The term "single-chain Fv" or "seFv" antibody refers to antibody
fragments
comprising the VH and VL domains of an antibody, wherein these domains are
present in a
single polypeptide chain. Generally, the Fv polypeptide further comprises a
polypeptide
linker between the VH and VL domains which enables the scFv to form the
desired structure
for antigen-binding. For a review of seFv, see Pluckthun (1994) THE
PHARMACOLOGY OF
MONOCLONAL ANTIBODIES, vol. 113, Rosenburg and Moore eds. Springer-Verlag, New
York, pp. 269-315. See also, International Patent Application Publication No.
WO 88/01649
and U.S. Pat. Nos. 4,946, 778 and 5,260,203.
-30-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
The present invention includes anti-TIGIT domain antibodies and methods of use
thereof. A "domain antibody" is an immunologically functional immunoglobulin
fragment
containing only the variable region of a heavy chain or the variable region of
a light chain. In
some instances, two or more VH regions are covalently joined with a peptide
linker to create a
bivalent domain antibody. The two VH regions of a bivalent domain antibody may
target the
same or different antigens.
The present invention includes anti-TIGIT bivalent antibodies and methods of
use
thereof. A "bivalent antibody" comprises two antigen-binding sites. In some
instances, the
two binding sites have the same antigen specificities. However, bivalent
antibodies may be
bispecific (see below).
The present invention includes anti-TIGIT diabodies and methods of use
thereof. As
used herein, the term "diabodies" refers to small antibody fragments with two
antigen-
binding sites, which fragments comprise a heavy chain variable domain (VH)
connected to a
light chain variable domain (VL) in the same polypeptide chain (VH-VL or VL-
VH). By using
a linker that is too short to allow pairing between the two domains on the
same chain, the
domains are forced to pair with the complemental.), domains of another chain
and create two
antigen-binding sites. Diabodies are described more fully in, e.g., EP
404,097; WO
93/11161; and Holliger et al. (1993) Proc. NatL Acad. S'ci. USA 90: 6444-6448.
For a review
of engineered antibody variants generally see Holliger and Hudson (2005) Nat.
Biotechnol.
23:1126-1136.
Typically, an antibody or antigen-binding fragment of the invention which is
modified
in some way retains at least 10% of its binding activity (when compared to the
parental
antibody) when that activity is expressed on a molar basis. Preferably, an
antibody or
antigen-binding fragment of the invention retains at least 20%, 50%, 70%, 80%,
90%, 95% or
100% or more of the TIGIT binding affinity as the parental antibody. It is
also intended that
an antibody or antigen-binding fragment of the invention can include
conservative or non-
conservative amino acid substitutions (referred to as "conservative variants"
or "function
conserved variants" of the antibody) that do not substantially alter its
biologic activity.
The present invention includes isolated anti-TIGIT antibodies and antigen-
binding
fragments thereof and methods of use thereof. "Isolated" antibodies or antigen-
binding
fragments thereof are at least partially free of other biological molecules
from the cells or cell
cultures in which they are produced. Such biological molecules include nucleic
acids,
proteins, lipids, carbohydrates, or other material such as cellular debris and
growth medium.
An isolated antibody or antigen-binding fragment may further be at least
partially free of
-31-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
expression system components such as biological molecules from a host cell or
of the growth
medium thereof. Generally, the term "isolated" is not intended to refer to a
complete absence
of such biological molecules or to an absence of water, buffers, or salts or
to components of a
pharmaceutical formulation that includes the antibodies or fragments.
The present invention includes monoclonal anti-TIGIT antibodies and antigen-
binding
fragments thereof as well as monoclonal compositions comprising a plurality of
isolated
monoclonal antibodies. The term "monoclonal antibody", as used herein, refers
to a
population of substantially homogeneous antibodies, i.e., the antibody
molecules comprising
the population are identical in amino acid sequence except for possible
naturally occurring
mutations that may be present in minor amounts. In contrast, conventional
(polyclonal)
antibody preparations typically include a multitude of different antibodies
having different
amino acid sequences in their variable domains, particularly their CDRs that
are often
specific for different epitopes. The modifier "monoclonal" indicates the
character of the
antibody as being obtained from a substantially homogeneous population of
antibodies, and is
not to be construed as requiring production of the antibody by any particular
method. For
example, the monoclonal antibodies to be used in accordance with the present
invention may
be made by the hybridoma method first described by Kohler et al. (1975) Nature
256: 495, or
may be made by recombinant DNA methods (see. e.g., U.S. Pat. No. 4,816,567).
The
"monoclonal antibodies" may also be isolated from phage antibody libraries
using the
techniques described in Clackson etal. (1991) Nature 352: 624-628 and Marks
etal. (1991)
J. Mol. Biol. 222: 581-597, for example. See also Presta (2005)J. Allergy
Glin. Immunol.
116:731.
The present invention includes anti-TIGIT chimeric antibodies (e.g., human
constant
domain/mouse variable domain) and methods of use thereof. As used herein, a
"chimeric
antibody" is an antibody having the variable domain from a first antibody and
the constant
domain from a second antibody, where the first and second antibodies are from
different
species. (U.S. Pat. No. 4,816,567; and Morrison etal., (1984) Proc. Natl.
Acad. Sci. USA 81:
6851-6855). Typically, the variable domains are obtained from an antibody from
an
experimental animal (the "parental antibody"), such as a rodent, and the
constant domain
sequences are obtained from human antibodies, so that the resulting chimeric
antibody will be
less likely to elicit an adverse immune response in a human subject than the
parental (e.g.,
mouse) antibody.
The present invention includes anti-TIGIT humanized antibodies and antigen-
binding
fragments thereof (e.g., rat or mouse antibodies that have been humanized) and
methods of
-32-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
use thereof. The invention includes any humanized version of the 14D7 antibody
(comprising SEQ ID NOs:7 and 8) or the 26B10 antibody (comprising SEQ ID
NOs:23 and
24). As used herein, the term "humanized antibody" refers to forms of
antibodies that contain
sequences from both human and non-human (e.g., mouse or rat) antibodies. In
general, the
humanized antibody will comprise substantially all of at least one, and
typically two, variable
domains, in which all or substantially all of the hypervariable loops
correspond to those of a
non-human immunoglobulin, and all or substantially all of the framework (FR)
regions are
those of a human immunoglobulin sequence. The humanized antibody may
optionally
comprise at least a portion of a human immunoglobulin constant region WO.
In general, the basic antibody structural unit comprises a tetramer. Each
tetramer
includes two identical pairs of polypeptide chains, each pair having one
"light" (about 25
kDa) and one "heavy" chain (about 50-70 kDa). The amino-terminal portion of
each chain
includes a variable region of about 100 to 110 or more amino acids primarily
responsible for
antigen recognition. The carboxy-terminal portion of the heavy chain may
define a constant
region primarily responsible for effector function. Typically, human light
chains are
classified as kappa and lambda light chains. Furthermore, human heavy chains
are typically
classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's
isotype as IgM,
IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the
variable and
constant regions are joined by a "J" region of about 12 or more amino acids,
with the heavy
chain also including a "D" region of about 10 more amino acids. See generally,
Fundamental
Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989).
The variable regions of each light/heavy chain pair form the antibody binding
site.
Thus, in general, an intact antibody has two binding sites. Except in
bifunctional or
bispecific antibodies, the two binding sites are, in general, the same.
Typically, the variable domains of both the heavy and light chains comprise
three
hypervariable regions, also called complementarity determining regions (CDRs),
located
within relatively conserved framework regions (FR). The CDRs are usually
aligned by the
framework regions, enabling binding to a specific epitope. In general, from N-
terminal to C-
terminal, both light and heavy chains variable domains comprise FR!. CDR1, FR2
, CDR2,
FR3, CDR3 and FR4. The assignment of amino acids to each domain is, generally,
in
accordance with the definitions of Sequences of Proteins of Immunological
Interest, Kabat, et
al.; National Institutes of Health, Bethesda, Md. ; 5th ed.; NIH Publ. No. 91-
3242 (1991);
Kabat (1978)Adv. Prot. Chem. 32:1-75; Kabat, etal., (1977)J. Biol. Chem.
252:6609-6616;
-33-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Chothia, etal., (1987) J Mol. Biol. 196:901-917 or Chothia, etal., (1989)
Nature 342:878-
883.
As used herein, the term "hypervariable region" refers to the amino acid
residues of an
antibody or antigen-binding fragment thereof that are responsible for antigen-
binding. The
hypervariable region comprises amino acid residues from a "complementarity
determining
region" or "CDR" (i.e. CDRL1, CDRL2 and CDRL3 in the light chain variable
domain and
CDRH1, CDRH2 and CDRH3 in the heavy chain variable domain). See Kabat etal.
(1991)
Sequences of Proteins of Immunological Interest, 5th Ed. Public Health
Service, National
Institutes of Health, Bethesda, Md. (defining the CDR regions of an antibody
by sequence);
see also Chothia and Lesk (1987) J Mol. Biol. 196: 901-917 (defining the CDR
regions of an
antibody by structure). As used herein, the term "framework" or "FR" residues
refers to those
variable domain residues other than the hypervariable region residues defined
herein as CDR
residues.
"Isolated nucleic acid molecule" or "isolated polynucleotide" means a DNA or
RNA
of genomic, mRNA, cDNA, or synthetic origin or some combination thereof which
is not
associated with all or a portion of a polynucleotide in which the isolated
polynucleotide is
found in nature, or is linked to a polynucleotide to which it is not linked in
nature. For
purposes of this disclosure, it should be understood that "a nucleic acid
molecule comprising"
a particular nucleotide sequence does not encompass intact chromosomes.
Isolated nucleic
acid molecules "comprising" specified nucleic acid sequences may include, in
addition to the
specified sequences, coding sequences for up to ten or even up to twenty or
more other
proteins or portions or fragments thereof, or may include operably linked
regulatory
sequences that control expression of the coding region of the recited nucleic
acid sequences,
andlor may include vector sequences.
The phrase "control sequences" refers to DNA sequences necessary for the
expression
of an operably linked coding sequence in a particular host organism. The
control sequences
that are suitable for prokaryotes, for example, include a promoter, optionally
an operator
sequence, and a ribosome binding site. Eukalyotic cells are known to use
promoters,
polyadenylation signals, and enhancers.
A nucleic acid or polynucleotide is "operably linked" when it is placed into a
functional relationship with another nucleic acid sequence. For example, DNA
for a
presequence or secretory leader is operably linked to DNA for a polypeptide if
it is expressed
as a preprotein that participates in the secretion of the polypeptide; a
promoter or enhancer is
operably linked to a coding sequence if it affects the transcription of the
sequence; or a
-34-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
ribosome binding site is operably linked to a coding sequence if it is
positioned so as to
facilitate translation. Generally, but not always, "operably linked" means
that the DNA
sequences being linked are contiguous, and, in the case of a secretory leader,
contiguous and
in reading phase. However, enhancers do not have to be contiguous. Linking is
accomplished by ligation at convenient restriction sites. If such sites do not
exist, the
synthetic oligonucleotide adaptors or linkers are used in accordance with
conventional
practice.
As used herein, the expressions "cell," "cell line," and "cell culture" are
used
interchangeably and all such designations include progeny. Thus, the words
"transformants"
and "transformed cells" include the primary subject cell and cultures derived
therefrom
without regard for the number of transfers. It is also understood that not all
progeny will
have precisely identical DNA content, due to deliberate or inadvertent
mutations. Mutant
progeny that have the same function or biological activity as screened for in
the originally
transformed cell are included. Where distinct designations are intended, it
will be clear from
the context.
As used herein, "germline sequence" refers to a sequence of unrearranged
immunoglobulin DNA sequences. Any suitable source of urirearranged
immunoglobulin
sequences may be used. Human germline sequences may be obtained, for example,
from
JOINSOLVER germline databases on the website for the National Institute of
Arthritis and
Musculoskeletal and Skin Diseases of the United States National Institutes of
Health. Mouse
germline sequences may be obtained, for example, as described in Giudicelli et
al. (2005)
Nucleic Acids Res. 33:D256-D261.
Physical and Functional Properties of the Exemplary Anti-TIGIT Antibodies
The present invention provides anti-TIGIT antibodies and antigen-binding
fragments
thereof having specified structural and functional features, and methods of
use of the
antibodies or antigen-binding fragments thereof in the treatment or prevention
of disease
(e.g., cancer or infectious disease).
An "anti-TIGIT antibody or antigen-binding fragment thereof of the present
invention" includes: any antibody or antigen-binding fragment thereof that is
discussed
herein (e.g., 14D7, 26B10 or humanized versions of these antibodies).
Cross-blocking antibodies and antigen-binding fragments thereof can be
identified
based on their ability to cross-compete with an antibody of the invention in
standard binding
assays (e.g., BIACore, ELBA, flow cls,,tometry). For example, standard ELISA
assays can be
-35-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
used in which a recombinant TIGIT (e.g., human TIGIT) protein is immobilized
on the plate,
one of the antibodies is fluorescently labeled and the ability of non-labeled
antibodies to
compete off the binding of the labeled antibody is evaluated. Additionally or
alternatively,
BlAcore analysis can be used to assess the ability of the antibodies to cross-
compete. The
ability of a test antibody to inhibit the binding of another antibody (for
example, antibody
14D7 or 26B10) to TIGIT (e.g., human TIGIT) demonstrates that the test
antibody can
compete with another antibody (e.g., 14D7 or 26B10) for binding to TIGIT
(e.g., human
TIGIT) and thus, may, in some cases, bind to the same epitope on TIGIT (e.g.,
human TIGIT)
as antibody 14D7 or 26B10 or to an overlapping epitope.
As stated above, antibodies and fragments that bind to the same epitope as any
of the
anti- TIGIT antibodies or antigen-binding fragments thereof of the present
invention also
form part of the present invention. Further, antibodies that bind to an
epitope that overlaps
with the epitope bound by any of the anti-TIGIT antibodies of the invention
also form part of
the present invention. There are several methods available for mapping
antibody epitopes on
target antigens, including: HID-Ex Mass spec, X-ray crystallography, pepscan
analysis and
site directed mutagenesis. For example, HDX (Hydrogen Deuterium Exchange)
coupled with
proteolysis and mass spectrometiy can be used to determine the epitope of an
antibody on a
specific antigen Y. HDX-MS relies on the accurate measurement and comparison
of the
degree of deuterium incorporation by an antigen when incubated in 1320 on its
own and in
presence of its antibody at various time intervals. Deuterium is exchanged
with hydrogen on
the amide backbone of the proteins in exposed areas whereas regions of the
antigen bound to
the antibody will be protected and will show less or no exchange after
analysis by LC-
MS/MS of proteolytic fragments.
Examples of the immunoglobulin chains of anti-TIGIT antibodies of the
invention as
well as their CDRs include, but are not limited those disclosed in Table 4.
The present
invention includes any polypeptide comprising or consisting of the amino acid
sequences of
SEQ ID NOs: 1-32, 45-112, and recombinant nucleotides encoding such
polypeptides.
The scope of the present invention includes isolated anti-TIGIT antibodies and

antigen-binding fragments thereof (e.g., humanized antibodies), comprising a
variant of an
immunoglobulin chain set forth herein, e.g., any of SEQ ID NOs:7-16, 48-56, 63-
80, 23-32,
and 89-112; wherein the variant exhibits one or more of the following
properties: (i) binds
human TIGIT; (ii) cross-reacts with cynomolgous and rhesus TIGIT; (iii) blocks
binding of
human TIGIT to human CD155 and human CD112; (iv) increases T cell activation;
(v)
-36-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
stimulates antigen-specific T-cell production of IL-2 and IFNT; (vi) blocks
induction of T cell
suppression of activation induced by TIGIT ligation with cognate ligands CD155
and CD112.
In other embodiments, the invention provides antibodies or antigen-binding
fragment
thereof that binds human TIGIT (e.g., humanized antibodies) and has VL domains
and VH
domains with at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence
identity with
SEQ ID NOs: 7-16, 48-56, 63-80 23-32, and 89-112; wherein the variant exhibits
the desired
binding and properties, e.g, (i) binds human TIGIT with a KD value of about 1
x 104 M to
about 1 x 1042 M as determined by surface plasmon resonance (e.g., BIACORE) or
a similar
technique (e.g. KinExa or OCTET); (ii) cross-reacts with cynomolgous and
rhesus TIGIT;
(iii) blocks binding of human TIGIT to human CD155 and human CD112; (iv)
increases T
cell activation; (v) stimulates antigen-specific T-cell production of IL-2 and
IFNI', (vi) blocks
induction of T cell suppression of activation induced by TIGIT ligation with
cognate ligands
CD155 and CD112.
"Conservatively modified variants" or "conservative substitution" refers to
substitutions of amino acids in a protein with other amino acids having
similar characteristics
(e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone
conformation and
rigidity, etc.), such that the changes can frequently be made without altering
the biological
activity of the protein. Those of skill in this art recognize that, in
general, single amino acid
substitutions in non-essential regions of a polypeptide do not substantially
alter biological
activity (see. e.g., Watson et al. (1987) Molecular Biology of/he Gene, The
Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of
structurally or
functionally similar amino acids are less likely to disrupt biological
activity. Exemplary
conservative substitutions are set forth in Table 1.
TABLE 1. Exemplary Conservative Amino Acid Substitutions
Ori_*nal residue Conservative substitution
"Iv; Ser
r_ R 31111
grANIIIIIIIIIMSer: Ala
EEUMIIIIaala.As Gin
MjZEIIIIIIIIIAsn; Gin
,eu (L) He: Val
-37-.

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Original residue Conservative substitution
Lys (K) Arg; His
Met (M) Leu; Ile; Tyr
Phe (F) Tyr; Met; Leu
Pro (P) Ala
Ser (S) \Thr
Thr (T) Ser
Trp (W) Tyr; Phe
Tyr (Y) Trp; Phe
Val (V) Ile; Leu
Function-conservative variants of the antibodies of the invention are also
contemplated by the present invention. "Function-conservative variants," as
used herein,
refers to antibodies or fragments in which one or more amino acid residues
have been
changed without altering a desired property, such an antigen affinity and/or
specificity. Such
variants include, but are not limited to, replacement of an amino acid with
one having similar
properties, such as the conservative amino acid substitutions of Table 1. Also
provided are
isolated polypeptides comprising the VL domains of the anti-TIGIT antibodies
of the
invention (e.g., SEQ ID NOs: 8, 13-16, 24, 29-33, 63-80), and isolated
polypeptides
comprising the VH domains of the anti-TIGIT antibodies of the invention (e.g.,
SEQ ID MN:
7, 9-12, 24, 26-29, 48-56, 89-112) having up to 0, 1, 2, 3, 4, 5, 6, 7, 8, 9,
10, 11, 12, 13, 14,
15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino
acid substitutions.
In another embodiment, provided is an antibody or antigen-binding fragment
thereof
that binds TIGIT and has VL domains and VH domains with at least 99% 98%, 97%,
96%,
95%, 90%, 85%, 80% or 75% sequence identity to one or more of the VI, domains
or VH
domains described herein, and exhibits specific binding to TIGIT. In another
embodiment
the binding antibody or antigen-binding fragment thereof of the present
invention comprises
VL and VH domains (with and without signal sequence) having up to 0, 1, 2, 3,
4, 5, 6, 7, 8, 9,
10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28,
29, 30 or more amino
acid substitutions, and exhibits specific binding to TIGIT.
Polynucleotides and Polypeptides
The present invention further comprises the polynucleotides encoding any of
the
polypeptides or immunoglobulin chains of anti-TIGIT antibodies and antigen-
binding
fragments thereof of the invention. For example, the present invention
includes the
polynucleotides encoding the amino acids described in SEQ ID NOs: 1-32 or 45-
112, as well
-38-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
as polynucleotides which hybridize thereto and, also, any polypeptide encoded
by such a
hybridizing polynucleotide.
In general, the polynucleotides hybridize under low, moderate or high
stringency
conditions, and encode antibodies or antigen-binding fragments thereof that
maintain the
ability to bind to TIGIT (human, rhesus and/or cynomolgous monkey, e.g.,
Macaca
fascicularis). A first polynucleotide molecule is "hybridizable" to a second
polynucleotide
molecule when a single stranded form of the first polynucleotide molecule can
anneal to the
second polynucleotide molecule under the appropriate conditions of temperature
and solution
ionic strength (see Sambrook, etal., supra). The conditions of temperature and
ionic strength
determine the "stringency" of the hybridization. Typical low stringency
hybridization
conditions include 55 C, 5X SSC, 0.1% SDS and no formamide; or 30% formamide,
5X
SSC, 0.5% SDS at 42 C. Typical moderate stringency hybridization conditions
are 40%
formamide, with 5X or 6X SSC and 0.1% SDS at 42 C. High stringency
hybridization
conditions are 50% formamide, 5X or 6X SSC at 42 C or, optionally, at a higher
temperature
(e.g., 57 C, 59 C, 60 C, 62 C, 63 C, 65 C or 68 C). In general, SSC is 0.15M
NaCl and
0.015M Na-citrate. Hybridization requires that the two polynucleotide contain
complementary sequences, although, depending on the stringency of the
hybridization,
mismatches between bases are possible. The appropriate stringency for
hybridizing
polynucleotides depends on the length of the polynucleotides and the degree of

complementation, variables well known in the art. The greater the degree of
similarity or
homology between two nucleotide sequences, the higher the stringency under
which the
nucleic acids may hybridize. For hybrids of greater than 100 nucleotides in
length, equations
for calculating the melting temperature have been derived (see Sambrook, el
al., supra, 9.50-
9.51). For hybridization with shorter polynucleotides, e.g., oligonucleotides,
the position of
mismatches becomes more important, and the length of the oligonucleotide
determines its
specificity (see Sambrook, etal., supra, 11.7-11.8).
In another embodiment, an isolated polynucleotide, for example DNA,
encoding the polypeptide chains of the isolated antibodies or antigen-binding
fragments set
forth herein is provided. In one embodiment, the isolated polynucleotide
encodes an
antibody or antigen-binding fragment thereof comprising at least one mature
immunoglobulin
light chain variable (VI) domain according to the invention and/or at least
one mature
immunoglobulin heavy chain variable (VH) domain according to the invention. In
some
embodiments the isolated polynucleotide encodes both a light chain and a heavy
chain on a
single polynucleotide molecule, and in other embodiments the light and heavy
chains are
-39-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
encoded on separate polynucleotide molecules. In another embodiment the
polynucleotides
further encodes a signal sequence.
In one embodiment, the invention comprises an isolated polynucleotide encoding
an
antibody heavy variable (VH) domain or an antigen-binding fragment thereof
comprising
CDR-I-11 (SEQ ID NO:1), CDR-H2 (SEQ ID NO:2) and CDR-H3 (SEQ ID NO:3, 45, 46,
or
47).
In one embodiment, the invention comprises an isolated polynucleotide encoding
an
antibody light chain variable (VL) domain or an antigen-binding fragment
thereof comprising
CDR-L1 (SEQ TD NO:4), CDR-L2 (SEQ ID NO:5) and CDR-L3 (SEQ ID NO:6, 57, 58,
59,
60, 61, or 62).
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VH) domain of SEQ ID NO: 7.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VL) domain of SEQ ID NO: 8.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VH) domain of any one of SEQ TD NOs: 9-
12.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VH) domain of any one of SEQ ID NOs: 48-
56.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VL) domain of any one of SEQ ID NOs: 13-
16.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VL) domain of any one of SEQ ID NOs: 63-
80.
In one embodiment, the invention comprises an isolated polynucleotide encoding
an
antibody heavy variable (VH) domain or an antigen-binding fragment thereof
comprising
CDR-H1 (SEQ ID NO:17), CDR-H2 (SEQ ID NO:18, 81, 82, 83, 84, 85, 86, 87, or
88) and
CDR-H3 (SEQ ID NO:19).
In one embodiment, the invention comprises an isolated polynucleotide encoding
an
antibody light chain variable (VL) domain or an antigen-binding fragment
thereof comprising
CDR-L1 (SEQ ID NO:20), CDR-L2 (SEQ ID NO:21) and CDR-L3 (SEQ ID NO:22).
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VH) domain of SEQ ID NO: 23.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VL) domain of SEQ ID NO: 24.
-40-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VII) domain of any one of SEQ TD NOs: 25-
28.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (W) domain of any one of SEQ ID NOs: 89-
112.
In one embodiment, the invention comprises an isolated polynucleotide encoding
the
immunoglobulin heavy chain variable (VI) domain of any one of SEQ ID NOs: 29-
32.
This present invention also provides vectors, e.g., expression vectors, such
as plasmids,
comprising the isolated polynucleotides of the invention, wherein the
polynucleotide is
operably linked to control sequences that are recognized by a host cell when
the host cell is
transfected with the vector. Also provided are host cells comprising a vector
of the present
invention and methods for producing the antibody or antigen-binding fragment
thereof or
polypeptide disclosed herein comprising culturing a host cell harboring an
expression vector
or a nucleic acid encoding the immunoglobulin chains of the antibody or
antigen-binding
fragment thereof in culture medium, and isolating the antigen or antigen-
binding fragment
thereof from the host cell or culture medium.
Also included in the present invention are polypeptides, e.g., immunoglobulin
polypeptides, comprising amino acid sequences that are at least about 75%
identical, 80%
identical, more preferably at least about 90% identical and most preferably at
least about 95%
identical (e.g., 95%, 96%, 97%, 98%, 99%, 100%) to the amino acid sequences of
the
antibodies provided herein when the comparison is performed by a BLAST
algorithm
wherein the parameters of the algorithm are selected to give the largest match
between the
respective sequences over the entire length of the respective reference
sequences (e.g. expect
threshold: 10; word size: 3; max matches in a quely range: 0; BLOSUM 62
matrix; gap costs:
existence 11, extension 1; conditional compositional score matrix adjustment).
Sequence identity refers to the degree to which the amino acids of two
polypeptides
are the same at equivalent positions when the two sequences are optimally
aligned.
The following references relate to BLAST algorithms often used for sequence
analysis: BLAST ALGORITHMS: Altschul et al. (2005) FEBS J. 272(20): 5101-5109;
Altschul, S.F., etal., (1990) J. MoL Biol. 215:403-410; Gish, W., etal.,
(1993) Nature Genet.
3:266-272; Madden, T.L., et aL , (1996) Meth. EnzyinoL 266:131-141; Altschul,
S.F., etal.,
(1997) Nucleic Acids Res. 25:3389-3402; Zhang, J., etal., (1997) Genome Res.
7:649-656;
Wootton, J.C., etal., (1993) Comput. Chem. 17:149-163; Hancock, J.M. etal.,
(1994)
Comput. App!. Biosci. 10:67-70; ALIGNMENT SCORING SYSTEMS: Dayhoff, M.O., et
al., "A model of evolutionary change in proteins." in Atlas of Protein
Sequence and Structure,
-41-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
(1978) vol. 5, suppl. 3. M.O. Dayhoff (ed.), pp. 345-352, Natl. Biomed. Res.
Found.,
Washington, DC; Schwartz, R.M., et al., "Matrices for detecting distant
relationships." in
Atlas of Protein Sequence and Structure, (1978) vol. 5, suppl. 3." M.O.
Dayhoff (ed.), pp.
353-358, Natl. Biomed. Res. Found, Washington, DC; Altschul, S.F., (1991) J.
Mol. Biol.
219:555-565; States, D.J., et al.,(1991)Methods 3:66-70; Henikoff, S., et al.,
(1992) Proc.
Natl. Acad. Sci. WA 89:10915-10919; Altschul, S.F., el al., (1993) J. Mol.
Evol. 36:290-300;
ALIGNMENT STATISTICS: Karlin, S., et al., (1990) Proc. Natl. Acad. Sci. USA
87:2264-
2268; Karlin, S., etal., (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877;
Dembo, A., etal.,
(1994) Ann. Prob. 22:2022-2039; and Altschul, S.F. "Evaluating the statistical
significance of
multiple distinct local alignments." in Theoretical and Computational Methods
in Genome
Research (S. Suhai, ed.), (1997) pp. 1-14, Plenum, New York.
Binding Affinity
By way of example, and not limitation, the antibodies and antigen-binding
fragments
disclosed herein may bind human TIGIT with a KD value of at least about 1 x
leM (i.e, a
KD value of 1 x 1 0-9 M or lower) as determined by surface plasmon resonance
(e.g.,
BIACORE) or a similar technique (e.g. KinExa or OCTET). In one embodiment, the

antibodies and antigen-binding fragments disclosed herein may bind human TIGIT
with a KD
value of at least about 1 x leM to about 1 x 1012M as determined by surface
plasmon
resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or OCTET). In
one
embodiment, the antibodies and antigen-binding fragments disclosed herein may
bind human
TIGIT with a K1) value of at about 1 x 10-9m to about 1 x 10"12M as determined
by surface
plasmon resonance (e.g., BIACORE) or a similar technique (e.g. KinExa or
OCTET). In one
embodiment, the antibodies and antigen-binding fragments disclosed herein may
bind human
TIGIT with a KD value of at least about 50 pM (i.e, a KD value of about 50 pM
or lower) as
determined by BIACORE or a similar technique. In one embodiment, the
antibodies and
antigen-binding fragments disclosed herein may bind human TIGIT with a KD
value of at
least about 10 pM (i.e, a KD value of about 10 pm lower) as determined by
BIACORE or a
similar technique. In one embodiment, the antibodies and antigen-binding
fragments of the
invention may bind to human TIGIT with a Kr, of about 50 pM to about 1 pM as
determined
by BIACORE or a similar technique.
-42-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Immune Cell Activation
In some embodiments, the antibodies or antigen binding fragments of the
invention
increase the activity of an immune cell. The increase of the activity of an
immune cell can be
detected using any method known in the art. In one embodiment, the increase in
activity of
an immune cell can be detected by measuring the proliferation of the immune
cell. For
example, an increase in activity of a T cell can be detected by measuring the
proliferation of
the T cell or signal transduction events such as tyrosine phosphorylation of
immune receptors
or downstream kinases that transmit signals to transcriptional regulators. In
other
embodiments, the increase in activity of an immune cell can be detected by
measuring CTL
or NK cell cytotoxic function on specific target cells or IFNy cytokine
responses, which are
associated with stimulation of anti-tumor immunity. In yet other embodiments,
the increase
in activity of an immune cell can be detected by measuring T cell activation
ex vivo in a
sample derived from the subject. In one embodiment, the increase in T cell
activity is
determined by: (i) measuring SEB (Staphylococcus Enterotoxin B) induced
production of
one or more pro-inflammatory cytokines selected from the group consisting of:
IL-2, TNFot,
IL-17, IFNy, IL-1(3, GM-CSF, RANTES, IL-6, 1L-8, IL-5 and IL-13; or (ii)
measuring mixed
lymphocyte reactions or direct anti-CD3 mAb stimulation of T cell receptor
(TCR) signaling
to induce production of a cytokine selected from the group consisting of: IL-
2, TNFot, IL-17,
IFNy, 1L-1(3, GM-CSF, RANTES, IL-6, IL-8, IL-5 and IL-13. In certain
embodiments, the
anti-TIGIT antibody or antigen binding fragment thereof of the present
invention will
stimulates antigen-specific T-cell production of IL-2 and/or IFNy by at least
1.5 fold.
The present invention includes antagonist anti-TIGIT antibodies and antigen-
binding
fragments thereof and methods of use thereof, e.g., humanized, antagonist anti-
TIGIT
antibodies and fragments. An antagonist anti-TIGIT antibody or antigen-binding
fragment
thereof antagonizes an activity of human TIGIT such as by inhibiting TIGIT
binding to
CD155 and CD112, and inhibiting functional ITIM signal transduction by TIGIT
upon
binding to CD155 and CD112. Measurement of anti-TIGIT antagonist activity can
be
assessed by demonstrating blocking of T cell suppression following TCR
activation induced
by TIGIT ligation with cognate ligands CD155 and CD112. Hence, in one
embodiment of
increased responses, treating with antagonist anti-'TIGIT antibodies are able
to rescue IL-2
responses to levels observed in T cells that are not repressed by CD155 or
CD112 induction
of TIGIT. In a more preferred level of activation, responses, following
treatment with an
anti-TIGIT antagonist antibody may increase responses to a level higher than T
cell responses
not repressed by CD155 or CD112.
-43-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Ability of Anti-hTIG1T Antibodies to Block Binding to hCD155 and hCD112
In some embodiments, the anti-TIGIT antibodies or antigen binding fragments of
the
invention are able to block binding of human MU to human CD155 and human
CD112.
The ability to block binding of human TIGIT to human CD155 and human CD112 can
be
determined using any method known in the art. In one embodiment, the ability
of the
antibodies to block binding of human TIGIT to human CD155 and human CD112 is
determined using an ELISA assay as described in Example 2.
Methods of Making Antibodies and Antigen-binding Fragments Thereof
Hybridoma cells that produce parental (e.g, rat or mouse) monoclonal anti-
TIGIT
antibodies or antigen-binding fragments thereof discussed herein may be
produced by
methods which are commonly known in the art. Such isolated hybridomas are part
of the
present invention. These methods include, but are not limited to, the
hybridoma technique
originally developed by Kohler, etal., (1975) (Nature 256:495-497), as well as
the trioma
technique (Bering, etal., (1988) Biomed. Biochim. Acta. 47:211-216 and
Hagiwara, etal.,
(1993) Hum. Antibod. Hybridomas 4:15), the human B-cell hybridoma technique
(Kozbor,
etal., (1983) Immunology Today 4:72 and Cote, etal., (1983) Proc. Natl. Acad.
Sci. U.S.A
80:2026-2030), the EBV-hybridoma technique (Cole, et al., in Monoclonal
Antibodies and
Cancer Therapy, Alan R. Liss, Inc., pp. 77-96, 1985), and electric field based
electrofusion
using a Cyto Pulse large chamber cull fusion electroporator (Cyto Pulse
Sciences, Inc., Glen
Burnie, MD). Preferably, mouse splenocytes are isolated and fused with PEG or
by
electrofusion to a mouse myeloma cell line based upon standard protocols. The
resulting
hybridomas may then be screened for the production of antigen-specific
antibodies. For
example, single cell suspensions of splenic lymphocytes from immunized mice
may by fused
to one-sixth the number of P3X63- Ag8.653 nonsecreting mouse myeloma cells
(ATCC,
CRL 1580) with 50% PEG. Cells may be plated at approximately 2 x 105 cells/mL
in a flat
bottom microtiter plate, followed by a two week incubation in selective medium
containing
20% fetal Clone Serum, 18% "653" conditioned media, 5% origen (IGEN), 4 mM L-
glutamine, 1 mM L-glutamine, 1 mM sodium pyruvate, 5mM HEPES, 0.055 mM 2-
mercaptoethanol, 50 units/ml penicillin, 50 mg/ml streptomycin, 50 mg/ml
gentamycin and
DC HAT (Sigma; the HAT is added 24 hours after the fusion). After two weeks,
cells may be
cultured in medium in which the HAT is replaced with HT. Individual wells may
then be
screened by ELISA for anti-'TIGIT monoclonal IgG antibodies. Once extensive
hybridoma
-44-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
growth occurs, medium can be observed usually after 10-14 days. The antibody
secreting
hybridomas may be replated, screened again, and if still positive for human
IgG, anti-TIGIT
monoclonal antibodies, can be subcloned at least twice by limiting dilution.
The stable
subclones may then be cultured in vitro to generate small amounts of antibody
in tissue
culture medium for characterization.
Thus, the present invention includes methods for making an anti-TIGIT antibody
or
antigen-binding fragment thereof of the present invention comprising culturing
a hybridoma
cell that expresses the antibody or fragment under condition favorable to such
expression and,
optionally, isolating the antibody or fragment from the hybridoma and/or the
growth medium
(e.g. cell culture medium).
The anti-TIGIT antibodies disclosed herein may also be produced recombinantly
(e.g., in an E co/ift7 expression system, a mammalian cell expression system
or a lower
eukaryote expression system). In this embodiment, nucleic acids encoding the
antibody
immunoglobulin molecules of the invention (e.g, VH or VI) may be inserted into
a pET-
based plasmid and expressed in the E. coli/T7 system. For example, the present
invention
includes methods for expressing an antibody or antigen-binding fragment
thereof or
immunoglobulin chain thereof in a host cell (e.g, bacterial host cell such as
E.coli such as
B1.21 or B1.21DE3) comprising expressing Ti RNA polymerase in the cell which
also
includes a polynucleotide encoding an immunoglobulin chain that is operably
linked to a T7
promoter. For example, in an embodiment of the invention, a bacterial host
cell, such as a E.
coli, includes a polynucleotide encoding the Ti RNA polymerase gene operably
linked to a
lac promoter and expression of the polymerase and the chain is induced by
incubation of the
host cell with IPTG (isopropyl-beta-D-thiogalactopyranoside).
There are several methods by which to produce recombinant antibodies which are
known in the art. One example of a method for recombinant production of
antibodies is
disclosed in U.S. Patent No. 4,816,567.
Transformation can be by any known method for introducing polynucleotides into
a
host cell. Methods for introduction of heterologous polynucleotides into
mammalian cells are
well known in the art and include dextran-mediated transfection, calcium
phosphate
precipitation, polybrene-mediated transfection, protoplast fusion,
electroporation,
encapsulation of the polynucleotide(s) in liposomes, biolistic injection and
direct
microinjection of the DNA into nuclei. In addition, nucleic acid molecules may
be
introduced into mammalian cells by viral vectors. Methods of transforming
cells are well
-45-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
known in the art. See, for example, U.S. Patent Nos. 4,399,216; 4,912,040;
4,740,461 and
4,959,455.
Thus, the present invention includes recombinant methods for making an anti-
TIGIT
antibody or antigen-binding fragment thereof of the present invention, or an
immunoglobulin
chain thereof, comprising introducing a polynucleotide encoding one or more
immunoglobulin chains of the antibody or fragment (e.g., heavy and/or light
immunoglobulin
chain); culturing the host cell (e.g.. CHO or Pichia or Pichia pastoris) under
condition
favorable to such expression and, optionally, isolating the antibody or
fragment or chain from
the host cell and/or medium in which the host cell is grown.
Anti-TIGIT antibodies can also be synthesized by any of the methods set forth
in U.S.
Patent No. 6,331,415.
Eukatyotic and prokaryotic host cells, including mammalian cells as hosts for
expression of the antibodies or fragments or immunoglobulin chains disclosed
herein are well
known in the art and include many immortalized cell lines available from the
American Type
Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary
(CHO) cells,
NSO, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney
cells (COS),
human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, 3T3 cells,
HEK-293 cells
and a number of other cell lines. Mammalian host cells include human, mouse,
rat, dog,
monkey, pig, goat, bovine, horse and hamster cells. Cell lines of particular
preference are
selected through determining which cell lines have high expression levels.
Other cell lines
that may be used are insect cell lines, such as SP) cells, amphibian cells,
bacterial cells, plant
cells and fungal cells. Fungal cells include yeast and filamentous fungus
cells including, for
example, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia
koclamae. Pichia
membranaefaciens, Pichia minuta (Ogataea minukz, Pichia lindneri), Pichia
opuntiae, Pichia
therm otolerans, Pichia salictaria, Pichia guercuum, Pichia Pichia stiptis,
Pichia
methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp.,
Hansenula
polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans,
Aspergillus
nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei,
Chrysosporium
lucknowense, Fusarium sp., Fusarium gramineum, Fusarium venenatum,
Physcomitrella
patens and Neurospora cra.ssa. Pichia sp., any Saccharomyces sp., Hansenula
polymorpha,
any Kluyveromyces sp., Candida albicans, any Aspergillus sp., Trichoderma
reesei,
Chrysosporium lucknowense, any Fusarium sp., Yarrowia hpolytica, and
Neurospora crassa.
When recombinant expression vectors encoding the heavy chain or antigen-
binding portion
or fragment thereof, the light chain and/or antigen-binding fragment thereof
are introduced
-46-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
into mammalian host cells, the antibodies are produced by culturing the host
cells for a period
of time sufficient to allow for expression of the antibody or fragment or
chain in the host cells
or secretion of the into the culture medium in which the host cells are grown.
Antibodies and antigen-binding fragments thereof and immunoglobulin chains can
be
recovered from the culture medium using standard protein purification methods.
Further,
expression of antibodies and antigen-binding fragments thereof and
immunoglobulin chains
of the invention (or other moieties therefrom) from production cell lines can
be enhanced
using a number of known techniques. For example, the glutamine synthetase gene
expression
system (the GS system) is a common approach for enhancing expression under
certain
conditions. The GS system is discussed in whole or part in connection with
European Patent
Nos. 0 216 846, 0 256 055, and 0 323 997 and European Patent Application No.
89303964.4.
Thus, in an embodiment of the invention, the mammalian host cells (e.g., CHO)
lack a
glutamine synthetase gene and are grown in the absence of glutamine in the
medium wherein,
however, the polynucleotide encoding the immunoglobulin chain comprises a
glutamine
synthetase gene which complements the lack of the gene in the host cell.
The present invention includes methods for purifying an anti-TIGIT antibody or
antigen-binding fragment thereof of the present invention comprising
introducing a sample
comprising the antibody or fragment to a purification medium (e.g., cation
exchange medium,
anion exchange medium, hydrophobic exchange medium, affinity purification
medium (e.g.,
protein-A, protein-G, protein-A/G, protein-L)) and either collecting purified
antibody or
fragment from the flow-through fraction of said sample that does not bind to
the medium; or,
discarding the flow-through fraction and eluting bound antibody or fragment
from the
medium and collecting the eluate. In an embodiment of the invention, the
medium is in a
column to which the sample is applied. In an embodiment of the invention, the
purification
method is conducted following recombinant expression of the antibody or
fragment in a host
cell, e.g., wherein the host cell is first lysed and, optionally, the lysate
is purified of insoluble
materials prior to purification on a medium.
In general, glycoproteins produced in a particular cell line or transgenic
animal will
have a glycosylation pattern that is characteristic for glycoproteins produced
in the cell line or
transgenic animal. Therefore, the particular glycosylation pattern of an
antibody will depend
on the particular cell line or transgenic animal used to produce the antibody.
However, all
antibodies encoded by the nucleic acid molecules provided herein, or
comprising the amino
acid sequences provided herein, comprise the instant invention, independent of
the
glycosylation pattern the antibodies may have. Similarly, in particular
embodiments,
-47-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
antibodies with a glycosylation pattern comprising only non-fucosylated N-
glycans may be
advantageous, because these antibodies have been shown to typically exhibit
more potent
efficacy than their fucosylated counterparts both in vitro and in vivo (See
for example,
Shinkawa et al., J. Biol. Chem. 278: 3466-3473 (2003); U.S. Patent Nos.
6,946,292 and
7,214,775). These antibodies with non-fucosylated N-glycans are not likely to
be
immunogenic because their carbohydrate structures are a normal component of
the
population that exists in human serum IgG.
The present invention includes polyclonal anti-TIGIT antibodies and antigen-
binding
fragments thereof, e.g., a composition comprising a plurality of anti-'TIGIT
antibodies and
fragments, which include one or more of the anti-TIGIT antibodies or antigen-
binding
fragments thereof of the present invention, and methods of use thereof. A
polyclonal
antibody is an antibody which was produced among or in the presence of one or
more other,
non-identical antibodies. In general, polyclonal antibodies are produced from
collections of
different B-lymphocytes, e.g., the B-lymphocyte of an animal treated with an
immunogen of
interest, which produces a population of different antibodies but which are
all directed to the
immunogen. Usually, polyclonal antibodies are obtained directly from an
immunized animal,
e.g., spleen, serum or ascites fluid.
The present invention includes bispecific and bifunctional antibodies and
antigen-
binding fragments having a binding specificity for TIGIT and another antigen
such as, for
example, PD-1 or PD-Li or LAG-3, and methods of use thereof. In an embodiment
of the
invention, the anti-TIGIT chains comprise any one of the VI-INL sequences
described in
Table 4, and the PD1 chains comprise the amino acid sequence of SEQ ID NOs: 36
and 37 or
of SEQ ID NOs: 38 and 39 (or an antigen binding fragment of any of said
sequences). A
bispecific or bifunctional antibody is an artificial hybrid antibody having
two different
heavy/light chain pairs and two different binding sites. Bispecific antibodies
can be produced
by a variety of methods including fusion of hybridomas or linking of Fab'
fragments. See,
e.g., Songsivilai, et al., (1990) Gin. Exp. Immunol. 79: 315-321, Kostelny,
etal., (1992) J
Immunol. 148:1547- 1553. In addition, bispecific antibodies may be formed as
"diabodies"
(Holliger, et al., (1993) PNAS USA 90:6444-6448) or as "Janusins" (Traunecker,
et al.,
(1991) F.MBO J. 10:3655-3659 and Traunecker, etal., (1992) Int. J. Cancer
Suppl. 7:51-52).
The present invention further includes anti-TIGIT antigen-binding fragments of
the
anti-TIGIT antibodies disclosed herein. The antibody fragments include F(ab)2
fragments,
which may be produced by enzymatic cleavage of an IgG by, for example, pepsin.
Fab
-48-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
fragments may be produced by, for example, reduction of F(ab)2 with
dithiothreitol or
mercaptoethylamine.
Immunoglobulins may be assigned to different classes depending on the amino
acid
sequences of the constant domain of their heavy chains. There are at least
five major classes
of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be
further
divided into subclasses (isotypes), e.g. IgGl, IgG2, IgG3 and IgG4; IgAl and
IgA2. The
invention comprises antibodies and antigen-binding fragments of any of these
classes or
subclasses of antibodies.
In one embodiment, the antibody or antigen-binding fragment comprises a heavy
chain constant region, e.g. a human constant region, such as yl, y2, y3, or y4
human heavy
chain constant region or a variant thereof. In another embodiment, the
antibody or antigen-
binding fragment comprises a light chain constant region, e.g. a human light
chain constant
region, such as lambda or kappa human light chain region or variant thereof.
By way of
example, and not limitation the human heavy chain constant region can be y4
and the human
light chain constant region can be kappa. In an alternative embodiment, the Fc
region of the
antibody is y4 with a Ser228Pro mutation (Schuurman, J et. al., Mot Immunol.
38: 1-8,
2001).
In one embodiment, the antibody or antigen-binding fragment comprises a heavy
chain constant region of the IgG1 subtype.
In some embodiments, different constant domains may be appended to humanized
VL
and VH regions derived from the CDRs provided herein. For example, if a
particular intended
use of an antibody (or fragment) of the present invention were to call for
altered effector
functions, a heavy chain constant domain other than human IgG1 may be used, or
hybrid
IgGllIgG4 may be utilized.
Although human IgG1 antibodies provide for long half-life and for effector
functions,
such as complement activation and antibody-dependent cellular cytotoxicity,
such activities
may not be desirable for all uses of the antibody. In such instances a human
IgG4 constant
domain, for example, may be used. The present invention includes anti-TIGIT
antibodies and
antigen-binding fragments thereof which comprise an IgG4 constant domain,
e.g., antagonist,
humanized anti-TTGIT antibodies and fragments, and methods of use thereof. In
one
embodiment, the IgG4 constant domain can differ from the native human IgG4
constant
domain (Swiss-Prot Accession No. P01861.1) at a position corresponding to
position 228 in
the EU system and position 241 in the KABAT system, where the native Ser108 is
replaced
-49-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
with Pro, in order to prevent a potential inter-chain disulfide bond between
Cys106 and
Cys109 (corresponding to positions Cys 226 and Cys 229 in the EU system and
positions Cys
239 and Cys 242 in the KABAT system) that could interfere with proper intra-
chain disulfide
bond formation. See Angal et at (1993) MoL Imunot 30:105. In other instances,
a modified
IgG1 constant domain which has been modified to increase half-life or reduce
effector
function can be used.
Antibody Engineering
Further included are embodiments in which the anti-MIT antibodies and
antigen-binding fragments thereof are engineered antibodies to include
modifications to
framework residues within the variable domains of a parental (e.g, mouse or
rat) monoclonal
antibody, e.g. to improve the properties of the antibody or fragment.
Typically, such
framework modifications are made to decrease the immunogenicity of the
antibody or
fragment. This is usually accomplished by replacing non-CDR residues in the
variable
domains (i.e. framework residues) in a parental (e.g. rodent) antibody or
fragment (the donor
antibody or fragment) with analogous residues from the immune repertoire of
the species in
which the antibody is to be used, e.g. human residues in the case of human
therapeutics (the
acceptor antibody). Such an antibody or fragment is referred to as a
"humanized" antibody or
fragment. In some cases it is desirable to increase or decrease the affinity,
or alter the
specificity of an engineered (e.g. humanized) antibody. The desired
immunoglobul in
characteristics can be achieved by the selection and combination of framework
residues from
the parental antibody and acceptor antibody. One approach is to mutate one or
more
framework residues in the parental antibody to the corresponding human
germline
sequence. More specifically, an antibody or fragment that has undergone
somatic mutation
can contain framework residues that differ from the germline sequence from
which the
antibody is derived. Such residues can be identified by comparing the parental
antibody or
fragment framework sequences to the human germline sequences from which the
antibody or
fragment is derived and mutating one or more framework residues in the
antibody to that of
the corresponding human germline sequence. Another approach is to revert
("backmutate")
to the original parental (e.g., rodent) residue at one or more positions of
the engineered (e.g.
humanized) antibody, e.g. to restore binding affinity that may have been lost
in the process of
replacing the framework residues. (See, e.g, U.S. Patent No. 5,693,762, U.S.
Patent No.
5,585,089, U.S. Patent No. 5,530,101, W09222653 and W09404679.)
-50-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
In certain embodiments, the anti-TIGIT antibodies and antigen-binding
fragments
thereof are engineered (e.g. humanized) to include modifications to in the
framework and/or
CDRs to improve their properties. Such engineered changes can be based on
molecular
modelling. A molecular model for the variable region for the parental (non-
human) antibody
sequence can be constructed to understand the structural features of the
antibody and used to
identify potential regions on the antibody that can interact with the antigen.
Conventional
CDRs are based on alignment of immunoglobulin sequences and identifying
variable regions.
Kabat et al., (1991) Sequences of Proteins of Immunological Interest, Kabat,
et al.; National
Institutes of Health, Bethesda. Md. ; 5th ed.; NTH Publ. No. 91-3242, Kabat
(1978) Adv. Prot.
Chem. 32:1-75; Kabat, etal., (1977)J. Biol. Chem. 252:6609-6616. Chothia and
coworkers
carefully examined conformations of the loops in crystal structures of
antibodies and
proposed hypervariable loops. Chothia, etal., (1987)J MoL Biol. 196:901-917 or
Chothia, et
al., (1989) Nature 342:878-883. There are variations between regions
classified as "CDRs"
and "hypervariable loops". Later studies (Raghunathan et al, (2012)J MO!
Recog. 25, 3,
103-113) analyzed several antibody ¨antigen crystal complexes and observed
that the antigen
binding regions in antibodies do not necessarily conform strictly to the "CDR"
residues or
"hypervarible" loops. The molecular model for the variable region of the non-
human
antibody can be used to guide the selection of regions that can potentially
bind to the antigen.
In practice the potential antigen binding regions based on model differ from
the conventional
"CDR"s or "hyper variable" loops. Commercial scientific software such as MOE
(Chemical
Computing Group) can be used for molecular modeling. Human frameworks can be
selected
based on best matches with the non-human sequence both in the frameworks and
in the
CDRs. For FR4 (framework 4) in VH, Vi regions for the human germlines are
compared
with the corresponding non-human region. In the case of FR4 (framework 4) in
VL, J-kappa
and J-Lambda regions of human germline sequences are compared with the
corresponding
non-human region. Once suitable human frameworks are identified, the CDRs are
grafted
into the selected human frameworks. In some cases certain residues in the VL-
VH interface
can be retained as in the non-human (parental) sequence. Molecular models can
also be used
for identifying residues that can potentially alter the CDR conformations and
hence binding
to antigen. In some cases, these residues are retained as in the non-human
(parental)
sequence. Molecular models can also be used to identify solvent exposed amino
acids that
can result in unwanted effects such as glycosylation, deamidation and
oxidation.
Developability filters can be introduced early on in the design stage to
eliminate/minimize
these potential problems.
-51-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Another type of framework modification involves mutating one or more residues
within the framework region, or even within one or more CDR regions, to remove
T cell
epitopes to thereby reduce the potential irnmunogenicity of the antibody. This
approach is
also referred to as "dennmunization" and is described in further detail in
U.S. Patent No.
7,125,689.
1.0 In particular embodiments, it will be desirable to change certain
amino acids
containing exposed side-chains to another amino acid residue in order to
provide for greater
chemical stability of the final antibody, so as to avoid deamidation or
isomerization. The
deamidation of asparagine may occur on NG, DG, NG, NS, NA, NT, QG or QS
sequences
and result in the creation of an isoaspartic acid residue that introduces a
kink into the
polypeptide chain and decreases its stability (isoaspartic acid effect).
Isomerization can occur
at DG, DS, DA or DT sequences. In certain embodiments, the antibodies of the
present
disclosure do not contain deamidation or asparagine isomerism sites.
For example, an asparagine (Asn) residue may be changed to Gln or Ala to
reduce the
potential for formation of isoaspartate at any Asn-Gly sequences, particularly
within a CDR.
A similar problem may occur at a Asp-Gly sequence. Reissner and Aswad (2003)
Cell. Mol.
Life Sc!. 60:1281. Isoaspartate formation may debilitate or completely
abrogate binding of an
antibody to its target antigen. See, Presta (2005)J Allergy Clin. Immunol.
116:731 at 734.
In one embodiment, the asparagine is changed to glutamine (Gin). It may also
be desirable to
alter an amino acid adjacent to an asparagine (Asn) or glutamine (Gin) residue
to reduce the
likelihood of deamidation, which occurs at greater rates when small amino
acids occur
adjacent to asparagine or glutamine. See, Bischoff & Kolbe (1994)J. Chromatog.
662:261.
In addition, any methionine residues (typically solvent exposed Met) or
nyptophane residues
in CDRs may be changed to Lys, Leu, Ala, or Phe or other amino acids in order
to reduce the
possibility that the methionine sulfur would oxidize, which could reduce
antigen-binding
affinity and also contribute to molecular heterogeneity in the final antibody
preparation. Id.
Additionally, in order to prevent or minimize potential scissile Asn-Pro
peptide bonds, it may
be desirable to alter any Asn-Pro combinations found in a CDR to (fin-Pro, Ala-
Pro, or Asn-
Ala. Antibodies with such substitutions are subsequently screened to ensure
that the
substitutions do not decrease the affinity or specificity of the antibody for
TIGIT, or other
desired biological activity to unacceptable levels.
-52-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
TABLE 2. Exemplary stabilizing CDR variants
CDR Residue Stabilizing Variant Sequence
Asn-Gly Gin-Gly, Ala-Gly, or Asn-Ala
(N-G) (Q-G), (A-G), or (N-A)
Asp-Gly Glu-Gly, Ala-Gly or Asp-Ala
(D-G) (E-G). (A-G), or (D-A)
Met (typically solvent exposed) Lys, Leu, Ala, or Phe
(M) (K), (L), (A), or (F)
Asn Gin or Ala
(N) (0) or (A)
Asn-Pro Gin-Pro, Ala-Pro, or Asn-Ala
(N-P) (Q-P), (A-P), or (N-A)
Antibody Engineering of the Fc region
The antibodies (e.g., humanized antibodies) and antigen-binding fragments
thereof
disclosed herein (e.g., 14D7, 26B10 and humanized versions thereof) can also
be engineered
to include modifications within the Fc region, typically to alter one or more
properties of the
antibody, such as serum half-life, complement fixation, Fc receptor binding,
and/or effector
function (e.g., antigen-dependent cellular cytotoxicity). Furthermore, the
antibodies and
antigen-binding fragments thereof disclosed herein (e.g, 14D7, 26B10 and
humanized
versions thereof) can be chemically modified (e.g, one or more chemical
moieties can be
attached to the antibody) or be modified to alter its glycosylation, again to
alter one or more
properties of the antibody or fragment. Each of these embodiments is described
in further
detail below. The numbering of residues in the Fc region is that of the EU
index of Kabat.
The antibodies and antigen-binding fragments thereof disclosed herein (e.g,
14D7,
26B10 and humanized versions thereof) also include antibodies and fragments
with modified
(or blocked) Fc regions to provide altered effector functions. See, e.g., U.S.
Pat. No.
5,624,821; W02003/086310; W02005/120571; W02006/0057702. Such modifications
can
be used to enhance or suppress various reactions of the immune system, with
possible
beneficial effects in diagnosis and therapy. Alterations of the Fc region
include amino acid
changes (substitutions, deletions and insertions), glycosylation or
deglycosylation, and adding
multiple Fc regions. Changes to the Fc can also alter the half-life of
antibodies in therapeutic
antibodies, enabling less frequent dosing and thus increased convenience and
decreased use
of material. See Presta (2005).1. Allergy Clin. Immunol. 116:731 at 734-35.
In one embodiment, the antibody or antigen-binding fragment of the invention
(e.g.,
14D7, 26B10 or humanized versions thereof) is an IgG4 isotype antibody or
fragment
comprising a Serine to Proline mutation at a position corresponding to
position 228 (S228P;
-53-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
EU index) in the hinge region of the heavy chain constant region. This
mutation has been
reported to abolish the heterogeneity of inter-heavy chain disulfide bridges
in the hinge
region (Angal etal. supra; position 241 is based on the Kabat numbering
system).
In one embodiment of the invention, the hinge region of CH1 is modified such
that
the number of cysteine residues in the hinge region is increased or decreased.
This approach
is described further in U.S. Patent No. 5,677,425. The number of cysteine
residues in the
hinge region of CH1 is altered, for example, to facilitate assembly of the
light and heavy
chains or to increase or decrease the stability of the antibody.
In another embodiment, the Fc hinge region of an antibody or antigen-binding
fragment of the invention (e.g., 14D7, 26B10 or humanized versions thereof) is
mutated to
decrease the biological half-life of the antibody or fragment. More
specifically, one or more
amino acid mutations are introduced into the CH2-CH3 domain interface region
of the Fc-
hinge fragment such that the antibody or fragment has impaired Staphylococcyl
protein A
(SpA) binding relative to native Fc-hinge domain SpA binding. This approach is
described in
further detail in U.S. Patent No. 6,165,745.
In another embodiment, the antibody or antigen-binding fragment of the
invention
(e.g., 14D7, 26B10 or a humanized version thereof) is modified to increase its
biological half-
life. Various approaches are possible. For example, one or more of the
following mutations
can be introduced: T252L, T2545, T256F, as described in U.S. Patent No.
6,277,375.
Alternatively, to increase the biological half-life, the antibody can be
altered within the CHI
or CL region to contain a salvage receptor binding epitope taken from two
loops of a CH2
domain of an Fc region of an IgG, as described in U.S. Patent Nos. 5,869,046
and 6,121,022.
In yet other embodiments, the Fc region is altered by replacing at least one
amino acid
residue with a different amino acid residue to alter the effector function(s)
of the antibody or
antigen-binding fragment. For example, one or more amino acids selected from
amino acid
residues 234, 235, 236, 237, 297, 318, 320 and 322 can be replaced with a
different amino
acid residue such that the antibody has an altered affinity for an effector
ligand and retains the
antigen-binding ability of the parent antibody. The effector ligand to which
affinity is altered
can be, for example, an Fc receptor or the Cl component of complement. This
approach is
described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260.
In another example, one or more amino acids selected from amino acid residues
329,
331 and 322 can be replaced with a different amino acid residue such that the
antibody has
altered Clq binding and/or reduced or abolished complement dependent
cytotoxicity (CDC).
This approach is described in further detail in U.S. Patent No. 6,194,551.
-54-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
In another example, one or more amino acid residues within amino acid
positions 231
and 239 are altered to thereby alter the ability of the antibody to fix
complement. This
approach is described further in PCT Publication WO 94/29351.
In yet another example, the Fc region is modified to decrease the ability of
the
antibody or antigen-binding fragment of the invention (e.g., 14D7, 26B10 or a
humanized
version thereof) to mediate antibody dependent cellular cytotoxicity (ADCC)
and/or to
decrease the affinity of the antibody or fragment for an Fcy receptor by
modifying one or
more amino acids at the following positions: 238, 239, 243, 248, 249, 252,
254, 255, 256,
258, 264, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289,
290, 292, 293,
294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326,
327, 329, 330,
331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398,
414, 416, 419,
430, 434, 435, 437, 438 or 439. This approach is described further in PCT
Publication WO
00/42072. Moreover, the binding sites on human IgG1 for FcyR1, FcyRIT, FcyRIII
and FcRn
have been mapped and variants with improved binding have been described (see
Shields et
aL (2001)1 Biol. chem. 276:6591-6604).
In one embodiment of the invention, the Fc region is modified to decrease the
ability
of the antibody of the invention (e.g., 14D7, 26B10 and humanized versions
thereof) to
mediate effector function and/or to increase anti-inflammatory properties by
modifying
residues 243 and 264. In one embodiment, the Fc region of the antibody or
fragment is
modified by changing the residues at positions 243 and 264 to alanine. In one
embodiment,
the Fc region is modified to decrease the ability of the antibody or fragment
to mediate
effector function and/or to increase anti-inflammatory properties by modifying
residues 243,
264, 267 and 328.
Effector Function Enhancement
In some embodiments, the Fc region of an anti-TIGIT antibody is modified to
increase the ability of the antibody or antigen-binding fragment to mediate
effector function
and/or to increase their binding to the Fcgamina receptors (FcyRs).
The term "Effector Function" as used herein is meant to refer to one or more
of
Antibody Dependant Cell mediated Cytotoxic activity (ADCC), Complement-
dependant
cytotoxic activity (CDC) mediated responses, Fc-mediated phagocytosis or
antibody
dependant cellular phagocytosis (ADCP) and antibody recycling via the FcRn
receptor.
The interaction between the constant region of an antigen binding protein and
various
Fc receptors (FcR) including FcgammaRI (CD64), FcgammaRII (CD32) and
FcgammaRIII
-55-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
(CD16) is believed to mediate the effector functions, such as ADCC and CDC, of
the antigen
binding protein. The Fc receptor is also important for antibody cross-linking,
which can be
important for anti-tumor immunity.
Effector function can be measured in a number of ways including for example
via
binding of the FcgammaRIII to Natural Killer cells or via FcgammaRI to
monocytes/macrophages to measure for ADCC effector function. For example an
antigen
binding protein of the present invention can be assessed for ADCC effector
function in a
Natural Killer cell assay. Examples of such assays can be found in Shields et
al, 2001 J. Biol.
Chem., Vol. 276, p 6591-6604; Chappel et al, 1993 J. Biol. Chem., Vol 268, p
25124-25131;
Lazar et al, 2006 PNAS, 103; 4005-4010.
The ADCC or CDC properties of antibodies of the present invention, or their
cross-
linking properties, may be enhanced in a number of ways.
Human IgG1 constant regions containing specific mutations or altered
glycosylation
on residue Asn297 have been shown to enhance binding to Fc receptors. In some
cases these
mutations have also been shown to enhance ADCC and CDC (Lazar et al. PNAS
2006, 103;
4005-4010; Shields et al. J Biol Chem 2001, 276; 6591-6604; Nechansky et al.
Mol Immunol,
2007, 44; 1815-1817).
In one embodiment of the present invention, such mutations are in one or more
of
positions selected from 239, 332 and 330 (IgG1), or the equivalent positions
in other IgG
isotypes. Examples of suitable mutations are S239D and 1332E and A330L. In one
embodiment, the antigen binding protein of the invention herein described is
mutated at
positions 239 and 332, for example S239D and 1332E or in a further embodiment
it is
mutated at three or more positions selected from 239 and 332 and 330, for
example S239D
and 1332E and A330L. (EU index numbering).
In an alternative embodiment of the present invention, there is provided an
antibody
comprising a heavy chain constant region with an altered glycosylation profile
such that the
antigen binding protein has enhanced effector function. For example, wherein
the antibody
has enhanced ADCC or enhanced CDC or wherein it has both enhanced ADCC and CDC

effector function. Examples of suitable methodologies to produce antigen
binding proteins
with an altered glycosylation profile are described in W02003011878,
W02006014679 and
EP1229125.
In a further aspect, the present invention provides "non-fucosylated" or
"afucosylated" antibodies. Non-fucosylated antibodies harbour a tri-mannosyl
core structure
of complex-type N-glycans of Fc without fucose residue. These glycoengineered
antibodies
-56-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
that lack core fucose residue from the Fc N-glycans may exhibit stronger ADCC
than
fucosylated equivalents due to enhancement of FcgammaRITTa binding capacity.
The present invention also provides a method for the production of an antibody
according to the invention comprising the steps of: a) culturing a recombinant
host cell
comprising an expression vector comprising the isolated nucleic acid as
described herein,
wherein the recombinant host cell does not comprise an alpha-1,6-
fucosyltransferase; and b)
recovering the antigen binding protein. The recombinant host cell may be not
normally
contain a gene encoding an alpha-1,6-fucosyltransferase (for example yeast
host cells such as
Pichia sp.) or may have been genetically modified to inactive an alpha-1,6-
fucosyltransferase.
Recombinant host cells which have been genetically modified to inactivate the
FUT8 gene
encoding an alpha-1,6-fucosyltransferase are available. See, e.g., the
POTELLIGENT"
technology system available from BioWa, Inc. (Princeton, N.J.) in which
CHOK1SV cells
lacking a functional copy of the FUT8 gene produce monoclonal antibodies
having enhanced
antibody dependent cell mediated cytotoxicity (ADCC) activity that is
increased relative to an
identical monoclonal antibody produced in a cell with a functional FUT8 gene.
Aspects of the
POTELLIGENTrm technology system are described in US7214775, US6946292,
W00061739 and W00231240. Those of ordinary skill in the art will also
recognize other
appropriate systems.
It will be apparent to those skilled in the art that such modifications may
not only be
used alone but may be used in combination with each other in order to further
enhance
effector function.
Production of Antibodies with Modified Glycosylation
In still another embodiment, the antibodies or antigen-binding fragments of
the
invention (e.g., 14D7, 26B10 and humanized versions thereof) comprise a
particular
glycosylation pattern. For example, an afucosylated or an aglycosylated
antibody or
fragment can be made (i.e., the antibody lacks fucose or glycosylation,
respectively). The
glycosylation pattern of an antibody or fragment may be altered to, for
example, increase the
affinity or avidity of the antibody or fragment for a TIGIT antigen. Such
modifications can
be accomplished by, for example, altering one or more of the glycosylation
sites within the
antibody or fragment sequence. For example, one or more amino acid
substitutions can be
made that result removal of one or more of the variable region framework
glycosylation sites
to thereby eliminate glycosylation at that site. Such aglycosylation may
increase the affinity
-57-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
or avidity of the antibody or fragment for antigen. See, e.g., U.S. Patent
Nos. 5,714,350 and
6,350,861.
Antibodies and antigen-binding fragments disclosed herein (e.g., 14D7, 26B10
and
humanized versions thereof) may further include those produced in lower
eukaryote host
cells, in particular fungal host cells such as yeast and filamentous fungi
have been genetically
engineered to produce glycoproteins that have mammalian- or human-like
glycosylation
patterns (See for example, Choi et at, (2003) Proc. Natl. Acad Sc!. 100: 5022-
5027;
Hamilton etal., (2003) Science 301: 1244-1246; Hamilton et al., (2006) Science
313: 1441-
1443; Nett et al., Yeast 28(3):237-52 (2011); Hamilton et al., Curr Opin
Biotechnol.
Oct:18(5):387-92 (2007)). A particular advantage of these genetically modified
host cells
over currently used mammalian cell lines is the ability to control the
glycosylation profile of
glycoproteins that are produced in the cells such that compositions of
glycoproteins can be
produced wherein a particular N-glycan structure predominates (see, e.g., U.S.
Patent No.
7,029,872 and U.S. Patent No. 7,449,308). These genetically modified host
cells have been
used to produce antibodies that have predominantly particular N-glycan
structures (See for
example, Li etal., (2006) Nat. Biotechnol. 24: 210-215).
In particular embodiments, the antibodies and antigen-binding fragments
thereof
disclosed herein (e.g, 14D7, 26B10 and humanized versions thereof) further
include those
produced in lower eukaryotic host cells and which comprise fucosylated and non-
fucosylated
hybrid and complex N-glycans, including bisected and multiantennary species,
including but
not limited to N-glycans such as GlcNAc(1.4)Man3G1cNAc2: Ga1(l.4)G1cNAc(I.
4)Man3G1cNAc2; NANA(l4)Gal(14)G1cNAc(14)Man3GIcNAc2.
In particular embodiments, the antibodies and antigen-binding fragments
thereof
provided herein (e.g, 14D7, 26B10 and humanized versions thereof) comprise
antibodies and
fragments comprising complex N-glycans, wherein at least 50%, 60%, 70%, 80%,
90%, 95%,
97%, 98%, 99%, or 100% of the complex N-gly cans in comprise the structure
NANA2Gal2G1cNAc2Man3G1cNAc2, wherein such structure is afucosylated. Such
structures
can be produced, e.g., in engineered Pichia pastoris host cells.
As used herein, the terms "N-glycan" and "glycoform" are used interchangeably
and
refer to an N-linked oligosaccharide, for example, one that is attached by an
asparagine-N-
acetylglucosamine linkage to an asparagine residue of a polypeptide. N-linked
glycoproteins
contain an N-acetylglucosamine residue linked to the amide nitrogen of an
asparagine residue
in the protein. The predominant sugars found on glycoproteins are glucose,
galactose,
mannose, fucose, N-acetylgalactosamine (GalNAc), N-acetylglucosamine (G1cNAc)
and
-58-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
sialic acid (e.g., N-acetyl-neuraminic acid (NANA)). The processing of the
sugar groups
occurs co-translationally in the lumen of the ER and continues post-
translationally in the
Golgi apparatus for N-linked glycoproteins.
N-glycans have a common pentasaccharide core of Man3G1cNAc2 ("Man" refers to
mannose; "Glc" refers to glucose; and "NAc" refers to N-acetyl; GIcNAc refers
to N-
acetylglucosamine). Usually, N-glycan structures are presented with the non-
reducing end to
the left and the reducing end to the right. The reducing end of the N-glycan
is the end that is
attached to the Asn residue comprising the glycosylation site on the protein.
N-glycans differ
with respect to the number of branches (antennae) comprising peripheral sugars
(e.g.,
GlcNAc, galactose, fucose and sialic acid) that are added to the Man3G1cNAc2
("Man3") core
structure which is also referred to as the "trimannose core", the
"pentasaccharide core" or the
"paucimannose core". N-glycans are classified according to their branched
constituents (e.g.,
high mannose, complex or hybrid). A "high mannose" type N-glycan has five or
more
mannose residues. A "complex" type N-glycan typically has at least one GlcNAc
attached to
the 1,3 mannose arm and at least one GlcNAc attached to the 1,6 mannose arm of
a
"trimannose" core. Complex N-glycans may also have galactose ("Gal") or N-
acetylgalactosamine ("GalNAc") residues that are optionally modified with
sialic acid or
derivatives (e.g, "NANA" or "NeuAc", where "Neu" refers to neuraminic acid and
"Ac"
refers to acetyl). Complex N-glycans may also have intrachain substitutions
comprising
"bisecting" G1cNAc and core fucose ("Fuc"). Complex N-glycans may also have
multiple
antennae on the "trimannose core," often referred to as "multiple antennary
glycans." A
"hybrid" N-glycan has at least one GlcNAc on the terminal of the 1,3 mannose
arm of the
trimannose core and zero or more mannoses on the 1,6 mannose arm of the
trimannose core.
The various N-glycans are also referred to as "glycoforms."
-59-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Antibody Physical Properties
The antibodies and antigen-binding fragments thereof disclosed herein (e.g.,
1414D7,
26B10 and humanized versions thereof) may further contain one or more
glycosylation sites
in either the light or heavy chain immunoglobulin variable region. Such
glycosylation sites
may result in increased immunogenicity of the antibody or fragment or an
alteration of the
pK of the antibody due to altered antigen-binding (Marshall etal. (1972) Annu
Rev Biochem
41:673-702; Gala and Morrison (2004)J Immunol 172:5489-94; Wallick eta!
(1988)J Exp
Med 168:1099-109; Spiro (2002) Glycobiology 12:43R-56R; Parekh et al (1985)
Nature
316:452-7; Mimura et al. (2000) Mol Immunol 37:697-706). Glycosylation has
been known
to occur at motifs containing an N-X-ST sequence.
is Each
antibody or antigen-binding fragment (e.g, 14D7, 26B10 or humanized versions
thereof) will have a unique isoelectric point (pI), which generally falls in
the pH range
between 6 and 9.5. The pI for an IgG1 antibody typically falls within the pH
range of 7-9.5
and the pl for an IgG4 antibody typically falls within the pH range of 6-8.
Each antibody or antigen-binding fragment (e.g., 14D7, 26B10 or humanized
versions
thereof) will have a characteristic melting temperature, with a higher melting
temperature
indicating greater overall stability in vivo (Krishnamurthy R and Manning MC
(2002) Curr
Pharm Biotechnol 3:361-71). In general, the Tmi (the temperature of initial
unfolding) may
be greater than 60 C, greater than 65 C, or greater than 70 C. The melting
point of an
antibody or fragment can be measured using differential scanning calorimetry
(Chen eta!
(2003) Pharm Res 20:1952-60; Ghirlando et al (1999) Immunol Lett 68:47-52) or
circular
dichroism (Murray etal. (2002)J. C'hromatogr Sci 40:343-9).
In a further embodiment, antibodies and antigen-binding fragments thereof
(e.g.,
14D7, 26B10 and humanized versions thereof) are selected that do not degrade
rapidly.
Degradation of an antibody or fragment can be measured using capillary
electrophoresis (CE)
and MALDI-MS (Alexander AJ and Hughes DE (1995) Anal Chem 67:3626-32).
In a further embodiment, antibodies (e.g., 14D7, 26B10 and humanized versions
thereof) and antigen-binding fragments thereof are selected that have minimal
aggregation
effects, which can lead to the triggering of an unwanted immune response
andlor altered or
unfavorable pharmacokinetic properties. Generally, antibodies and fragments
are acceptable
with aggregation of 25% or less, 20% or less, 15% or less, 10% or less, or 5%
or less.
Aggregation can be measured by several techniques, including size-exclusion
column (SEC),
high performance liquid chromatography (HPLC), and light scattering.
-60-

CA 02994555 2018-02-01
WO 2017/030823 PCT/US2016/046100
Antibody Conjugates
The anti-'TIGIT antibodies and antigen-binding fragments thereof disclosed
herein
(e.g., 14D7, 26B10 and humanized versions thereof) may also be conjugated to a
chemical
moiety. The chemical moiety may be, inter alia, a polymer, a radionuclide or a
cytotoxic
factor. In particular embodiments, the chemical moiety is a polymer which
increases the
half-life of the antibody or fragment in the body of a subject. Suitable
polymers include, but
are not limited to, hydrophilic polymers which include but are not limited to
polyethylene
glycol (PEG) (e.g., PEG with a molecular weight of 2kDa, 5 kDa, 10 kDa, 12kDa,
20 kDa,
30kDa or 40kDa), dextran and monomethoxypolyethylene glycol (mPEG). Lee, et
al., (1999)
(Bioconj. Chem. 10:973-981) discloses PEG conjugated single-chain antibodies.
Wen, etal.,
(2001) (Bioconj Chem. 12:545-553) disclose conjugating antibodies with PEG
which is
attached to a radiometal chelator (diethylenetriaminpentaacetic acid (DTPA)).
The antibodies and antigen-binding fragments thereof disclosed herein (e.g.,
14D7,
26B10 and humanized versions thereof) may also be conjugated with labels such
as 99Tc,90Y,
"In, "P, I4c, 1251, 3H, 1311, 11c, 15 -
0 "N, 18F, 35S, "Cr, "To, 226Ra, 60 -0,
C 59Fe, 57Se, I52EU,
67cu, 2'7ci, 211At, 212ph, 47s0, 109pd, 234Th, and 40¨,
K Thd, "mn, 52Tr, and 56Fe.
The antibodies and antigen-binding fragments disclosed herein (e.g., 14D7,
26B10
and humanized versions thereof) may also be PEGylated, for example to increase
its
biological (e.g., serum) half-life. To PEGylate an antibody or fragment, the
antibody or
fragment, typically is reacted with a reactive form of polyethylene glycol
(PEG), such as a
reactive ester or aldehyde derivative of PEG, under conditions in which one or
more PEG
groups become attached to the antibody or antibody fragment. In particular
embodiments,
the PEGylation is carried out via an acylation reaction or an akilation
reaction with a
reactive PEG molecule (or an analogous reactive water-soluble polymer). As
used herein, the
term "polyethylene glycol" is intended to encompass any of the forms of PEG
that have been
used to derivatize other proteins, such as mono (CI-CIO) alkov- or aryloxy -
polyethylene
glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody
or fragment
to be PEGylated is an aglycosylated antibody or fragment. Methods for
PEGylating proteins
are known in the art and can be applied to the antibodies of the invention.
See, e.g., EP 0 154
316 and EP 0 401 384.
The antibodies and antigen-binding fragments disclosed herein (e.g., 14D7,
26B10
and humanized versions thereof) may also be conjugated with fluorescent or
chemilltuninescent labels, including fluorophores such as rare earth chelates,
fluorescein and
its derivatives, rhodamine and its derivatives, isothiocyanate, phycoetythrin,
phycocyanin,
-61-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
allophy,rcocyanin, o-phthaladehyde, fluorescamine, 152Eu, dansyl,
umbelliferone, luciferin,
luminal label, isoluminal label, an aromatic acridinium ester label, an
imidazole label, an
acridimium salt label, an oxalate ester label, an aequorin label, 2,3-
dihydrophthalazinediones.
biotinlavidin, spin labels and stable free radicals.
The antibodies and antigen-binding fragments thereof of the invention (e.g.,
14D7,
26B10 and humanized versions thereof) may also be conjugated to a cytotoxic
factor such as
diptheria toxin, Pseudomonas aeruginosa exotoxin A chain , ricin A chain,
abrin A chain;
modeccin A chain, alpha-sarcin, Aleurites fordii proteins and compounds (e.g.,
fatty acids),
dianthin proteins, Phytoiacca americana proteins PAP!, PAPII, and PAP-S,
momordica
charantia inhibitor, curcin, crotin, saponaria officincilis inhibitor,
mitogellin, restrictocin,
is phenomycin, and enomycin.
Any method known in the art for conjugating the antibodies and antigen-binding

fragments thereof of the invention (e.g., 14D7, 26B10 and humanized versions
thereof) to the
various moieties may be employed, including those methods described by Hunter,
et al.,
(1962) Nature 144:945; David, et al., (1974) Biochemistry 13:1014; Pain, et
al., (1981) J.
Immunol. Meth. 40:219; and Nygren, J., (1982) Hislochem. and Cytochem. 30:407.
Methods
for conjugating antibodies and fragments are conventional and very well known
in the art.
Therapeutic Uses of Anti-TIGIT antibodies
Further provided are methods for treating subjects, including human subjects,
in need
of treatment with the isolated antibodies or antigen-binding fragments thereof
disclosed
herein (e.g., 14D7, 26B10 and humanized versions thereof). In one embodiment
of the
invention, such subject suffers from an infection or an infectious disease. In
another
embodiment of the invention, such subject suffers from cancer. In one
embodiment, the
cancer is a solid tumor which is infiltrated by tumor-infiltrating lymphocytes
which express
TIGIT. In one embodiment the cancer is , e.g., osteosarcoma, rhabdomyosarcoma,
neuroblastoma, kidney cancer, leukemia, renal transitional cell cancer,
bladder cancer,
Wilm's cancer, ovarian cancer, pancreatic cancer, breast cancer, prostate
cancer, bone cancer,
lung cancer (e.g., non-small cell lung cancer), gastric cancer, colorectal
cancer, cervical
cancer, synovial sarcoma, head and neck cancer, squatnous cell carcinoma,
multiple
myeloma, renal cell cancer, retinoblastoma, hepatoblastoma, hepatocellular
carcinoma,
melanoma, rhabdoid tumor of the kidney. Ewing's sarcoma, chondrosarcoma, brain
cancer,
glioblastoma, meningioma, pituitary adenoma, vestibular schwannoma, a
primitive
neuroectodermal tumor. medulloblastoma, astrocytoma, anaplastic astrocytoma,
-62-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
oligodendroglioma, ependymoma, choroid plexus papilloma, polycythemia vera,
thrombocythemia, idiopathic myelfibrosis, soft tissue sarcoma, thyroid cancer,
endometrial
cancer, carcinoid cancer or liver cancer, breast cancer or gastric cancer. In
an embodiment of
the invention, the cancer is metastatic cancer, e.g, of the varieties
described above.
In an embodiment, the invention provides methods for treating subjects using
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 and
humanized versions thereof), wherein the subject suffers from a viral
infection. In one
embodiment, the viral infection is infection with a virus selected from the
group consisting of
human immunodeficiency virus (HIV), hepatitis virus (A, B, or C), herpes virus
(e.g., VZV,
HSV-I, HAV-6, HSV-II, and CMV, Epstein Barr virus), adenovirus, influenza
virus,
flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory
syncytial virus,
mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia
virus, HTLV virus,
dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC
virus or arboviral
encephalitis virus.
In an embodiment, the invention provides methods for treating subjects using
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention, wherein
the subject
suffers from a bacterial infection. In one embodiment, the bacterial infection
is infection with
a bacteria selected from the group consisting of Chlamydia, rickettsial
bacteria, rnycobacteria,
staphylococci, streptococci, pneumonococci, meningococci and gonococci,
klebsiella,
proteus, serratia, pseudomonas, Legionella, Corynebacierium diphtheriae,
Salmonella,
bacilli, Vibrio cholerae, Clostridium tetan, Clostridium botulinum, Bacillus
anthricis,
Yersinia pest/s. Mycobacterium leprae, Mycobacterium lepromatosis, and
Borriella.
In an embodiment, the invention provides methods for treating subjects using
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention, wherein
the subject
suffers from a fungal infection. In one embodiment, the fungal infection is
infection with a
fungus selected from the group consisting of Candida (albicans, krusei,
glabrata, tropicalis,
etc.), Cryptococcus neofirmans, Aspergillus (fumigatus, niger, etc.), Genus
Mucorales
(mucor, absidia, rhizopus), Sporothrix schenkii, Blastomyces dermatitidis,
Paracoccidioides
brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
In an embodiment, the invention provides methods for treating subjects using
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention, wherein
the subject
suffers from a parasitic infection. In one embodiment, the parasitic infection
is infection with
a parasite selected from the group consisting of Entamoeba histolytica,
Balantidium con,
Naegleria fowleri, Acanihamoeba, Giardia lamb/a, Cryptosporidium, Pneumocystis
car/nil,
-63-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi,
Leishmania
donovani, Toxoplasma gondii and Nippostrongylus brasiliensis.
In addition, the present invention provides a method for preventing or
inhibiting
TIGIT binding to MHC class II, enhancing antigen-specific T-cell activation or
stimulating
T-cell production of interleukin-2 in a subject (e.g., human), for example,
wherein the subject
suffers from cancer or infectious disease (e.g., as discussed herein)
comprising administering
an effective amount of anti-TIGIT antibody or antigen-binding fragment thereof
(e.g, 14D7,
26B10 and humanized versions thereof), optionally in association with a
further
chemotherapeutic agent.
A "subject" may be a mammal such as a human, dog, cat, horse, cow, mouse, rat,
monkey (e.g, cynomolgous monkey, e.g.. Macaca fascicularis) or rabbit. In
preferred
embodiments of the invention, the subject is a human subject.
In particular embodiments, the antibodies or antigen-binding fragments thereof

disclosed herein (e.g., 14D7, 26B10 and humanized versions thereof) may be
used alone, or
in association with other, further therapeutic agents and/or therapeutic
procedures, for
treating or preventing any disease such as cancer, e.g., as discussed herein,
in a subject in
need of such treatment or prevention. Compositions, e.g., pharmaceutical
compositions
comprising a pharmaceutically acceptable carrier, comprising such antibodies
and fragments
in association with further therapeutic agents are also part of the present
invention.
In particular embodiments, the antibodies or antigen-binding fragments thereof
disclosed herein (e.g., 14D7, 26B10 and humanized versions thereof) may be
used alone, or
in association with tumor vaccines.
In particular embodiments, the antibodies or antigen-binding fragments thereof

disclosed herein (e.g , 14D7, 26B10 and humanized versions thereof) may be
used alone, or
in association with chemotherapeutic agents.
In particular embodiments, the antibodies or antigen-binding fragments thereof
disclosed herein (e.g., 14D7, 26B10 and humanized versions thereof) may be
used alone, or
in association with radiation therapy.
In particular embodiments, the antibodies or antigen-binding fragments thereof

disclosed herein (e.g., 14D7, 26B10 and humanized versions thereof) may be
used alone, or
in association with targeted therapies. Examples of targeted therapies
include: hormone
therapies, signal transduction inhibitors (e.g., EGFR inhibitors, such as
cetuximab (Erbitux)
and erlotinib (Tarceva)); HER2 inhibitors (e.g., trastuzumab (Herceptin) and
pertuzumab
(Perjeta)); BCR-ABL inhibitors (such as imatinib (Gleevec) and dasatinib
(Sprycel)); ALK
-64-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
inhibitors (such as crizotinib (Xalkori) and ceritinib (Zykadia)); BRAF
inhibitors (such as
vemurafenib (Zelboraf) and dabrafenib (Tafinlar)), gene expression modulators,
apoptosis
inducers (e.g., bortezomib (Velcade) and carfilzomib (Kyprolis)), angiogenesis
inhibitors
(e.g., bevacizumab (Avastin) and ramucirumab (Cyramza) , monoclonal antibodies
attached
to toxins (e.g., brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine
(Kadcyla)).
1.0 In particular embodiments, the anti-TIGIT antibodies or antigen-binding
fragments
thereof of the invention (e.g, 14D7, 26B10 and humanized versions thereof) may
be used in
combination with an anti-cancer therapeutic agent or immunomodulatory drug
such as an
immunomodulatoty receptor inhibitor, e.g., an antibody or antigen-binding
fragment thereof
that specifically binds to the receptor.
Thus, the present invention includes compositions comprising an anti-TIGIT
antibody
or antigen-binding fragment thereof of the present invention (e.g., 14D7,
26B10 and
humanized versions thereof) in association with pembrolizumab; as well as
methods for
treating or preventing cancer in a subject comprising administering an
effective amount of the
anti-TIGIT antibody or antigen-binding fragment thereof and pembrolizumab to
the subject.
Optionally, the subject is also administered a further therapeutic agent.
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the present invention (e.g., 14D7, 26B10 and humanized
versions
thereof) is in association with an isolated antibody comprising an
immunoglobulin heavy
chain comprising the amino acid sequence of SEQ TD NO: 36 and an
immunoglobulin light
chain comprising the amino acid sequence of SEQ ID NO: 37. SEQ ID NOs: 36 and
37
encode the heavy and light chain of pembrolizumab.
In an embodiment of the invention, an anti-TIM' antibody ) or antigen-binding
fragment thereof of the present invention (e.g., 14D7, 26B10 and humanized
versions
thereof) is in association with an isolated antibody comprising an
immunoglobulin heavy
chain comprising the amino acid sequence of SEQ ID NO: 38 and an
immunoglobulin light
chain comprising the amino acid sequence of SEQ TD NO: 39. SEQ ID NOs: 38 and
39
encode the heavy and light chain of nivolumab.
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g., I 4D7, 26B10 and humanized versions
thereof) is in
association with one or more of: anti-PD! antibody (e.g., pembrolizumab,
nivolumab,
pidilizumab (CT-011)), anti-PDL1 antibody, anti-CTLA4 antibody, anti-CS1
antibody (e.g.,
elotuzumab), anti-KIR2DL1/2/3 antibody (e.g., lirilumab), anti-CD137 antibody
(e.g.,
urelumab), anti-GITR antibody (e.g., TRX518), anti-PD-L1 antibody (e.g., BMS-
936559,
-65-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
MSB0010718C or MPDL3280A), anti-PD-L2 antibody, anti-ILT1 antibody, anti-ILT2
antibody, anti-ILT3 antibody, anti-ILT4 antibody, anti-ILT5 antibody, anti-
ILT6 antibody,
anti-ILT7 antibody, anti-ILT8 antibody, anti-CD40 antibody, anti-0X40
antibody, anti-
ICOS, anti-SIRPa, anti-KIR2DL1 antibody, anti-KIR2DL2/3 antibody, anti-KIR2DL4

antibody, anti-KIR2DL5A antibody, anti-KIR2DL5B antibody, anti-KIR3DL1
antibody,
anti-KIR3DL2 antibody, anti-KIR3DL3 antibody, anti-NKG2A antibody, anti-NKG2C
antibody, anti-NKG2E antibody, anti-4-1BB antibody (e.g., PP-05082566), anti-
TSLP
antibody, anti-IL-10 antibody, IL-10 or PEGylated IL-10, or any small organic
molecule
inhibitor of such targets.
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g, 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-PD1 antibody. In an embodiment of the invention, an
anti-TIGIT
antibody or antigen-binding fragment thereof of the invention (e.g., 14D7,
26B10 or
humanized versions thereof) is in association with an anti-PDL1 antibody
(e.g., BMS-
936559, MSB0010718C or MPDL3280A). In an embodiment of the invention, an anti-
TTGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 or
humanized versions thereof) is in association with an anti-CTLA4 antibody. In
an
embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of
the invention (e.g., 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-CS1 antibody. In an embodiment of the invention, an anti-TIGIT antibody
or antigen-
binding fragment thereof of the invention (e.g., 14D7, 26B10 or humanized
versions thereof)
is in association with an anti-KIR2DL1/2/3 antibody. In an embodiment of the
invention, an
anti-TIGTT antibody or antigen-binding fragment thereof of the invention
(e.g., 14D7, 26B10
or humanized versions thereof) is in association with an anti-CD137 (e.g.,
ureltunab)
antibody. In an embodiment of the invention, an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-GITR (e.g.. TRX518) antibody. In an embodiment of the
invention,
an anti-TIGIT antibody or antigen-binding fragment thereof of the invention
(e.g., 14D7,
26B10 or humanized versions thereof) is in association with an anti-PD-L2
antibody. In an
embodiment of the invention, an anti-TIGTT antibody or antigen-binding
fragment thereof of
the invention (e.g., 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-ITL1 antibody. In an embodiment of the invention, an anti-TIGIT antibody
or antigen-
binding fragment thereof of the invention (e.g., 14D7, 26B10 or humanized
versions thereof)
is in association with an anti-ITL2 antibody. In an embodiment of the
invention, an anti-
-66-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 or
humanized versions thereof) is in association with an anti-ITL3 antibody. In
an embodiment
of the invention, an anti-TIGIT antibody or antigen-binding fragment thereof
of the invention
(e.g., 14D7, 26B10 or humanized versions thereof) is in association with an
anti-1TL4
antibody. In an embodiment of the invention, an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-ITL5 antibody. In an embodiment of the invention, an
anti-TIGIT
antibody or antigen-binding fragment thereof of the invention (e.g., 14D7,
26B10 or
humanized versions thereof) is in association with an anti-ITL6 antibody. In
an embodiment
of the invention, an anti-TIGIT antibody or antigen-binding fragment thereof
of the invention
(e.g., 14D7, 26B10 or humanized versions thereof) is in association with an
anti-1TL7
antibody. In an embodiment of the invention, an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-ITL8 antibody. In an embodiment of the invention, an
anti-TIGIT
antibody or antigen-binding fragment thereof of the invention (e.g., 14D7,
26B10 or
humanized versions thereof) is in association with an anti-CD40 antibody. In
an embodiment
of the invention, an anti-TIGIT antibody or antigen-binding fragment thereof
of the invention
(e.g., 14D7, 26B10 or humanized versions thereof) is in association with an
anti-0X40
antibody. In an embodiment of the invention, an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-KIR2DLI antibody. In an embodiment of the invention,
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 or
humanized versions thereof) is in association with an anti-KIR2DL2/3 antibody.
In an
embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of
the invention (e.g, 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-KIR2DL4 antibody. In an embodiment of the invention, an anti-TIGIT
antibody or
antigen-binding fragment thereof of the invention (e.g., 14D7, 26B10 or
humanized versions
thereof) is in association with an anti-KIR2DL5A antibody. In an embodiment of
the
invention, an anti-TIGIT antibody or antigen-binding fragment thereof of the
invention (e.g.,
14D7, 26B10 or humanized versions thereof) is in association with an anti-
KIR2DL5B
antibody. In an embodiment of the invention, an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention (e.g, 14D7, 26B10 or humanized versions
thereof) is in
association with an anti-KIR3DL1 antibody. In an embodiment of the invention,
an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 or
-67-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
humanized versions thereof) is in association with an anti-KIR3DL2 antibody.
In an
embodiment of the invention, an anti-TIGTT antibody or antigen-binding
fragment thereof of
the invention (e.g, 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-KIR3DL3 antibody. In an embodiment of the invention, an anti-TIGIT
antibody or
antigen-binding fragment thereof of the invention (e.g., 14D7, 26B10 or
humanized versions
thereof) is in association with an anti-NKG2A antibody. In an embodiment of
the invention,
an anti-TIGIT antibody or antigen-binding fragment thereof of the invention
(e.g, 14D7,
26B10 or humanized versions thereof) is in association with an anti-NKG2C
antibody. In an
embodiment of the invention, an anti-TIGTT antibody or antigen-binding
fragment thereof of
the invention (e.g, 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-ICOS antibody. In an embodiment of the invention, an anti-TIGIT antibody
or antigen-
binding fragment thereof of the invention (e.g., 14D7, 26B10 or humanized
versions thereof)
is in association with an anti-SIRPa antibody. In an embodiment of the
invention, an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
14D7, 26B10 or
humanized versions thereof) is in association with an anti-4-1BB antibody. In
an
embodiment of the invention, an anti-TIGTT antibody or antigen-binding
fragment thereof of
the invention (e.g, 14D7, 26B10 or humanized versions thereof) is in
association with an
anti-IL-10 antibody. In an embodiment of the invention, an anti-TIGIT antibody
or antigen-
binding fragment thereof of the invention (e.g., 14D7, 26B10 or humanized
versions thereof)
is in association with an anti-TSLP antibody. In an embodiment of the
invention, an anti-
TIGIT antibody or antigen-binding fragment thereof of the invention (e.g.,
I4D7, 26B10 or
humanized versions thereof) is in association with IL-10 or PEGylated IL-10.
In an embodiment of the invention, an anti-TIGTT antibody or antigen-binding
fragment thereof of the invention (e.g, 14D7, 26B10 or humanized versions
thereof) is in
association with one or more of an inhibitor (e.g, a small organic molecule or
an antibody or
antigen-binding fragment thereof) such as: an MTOR (mammalian target of
rapamycin)
inhibitor, a cls,,totoxic agent, a platinum agent, an EGFR inhibitor, a VEGF
inhibitor, a
microtubule stabilizer, a taxane, a CD20 inhibitor, a CD52 inhibitor, a CD30
inhibitor, a
RANK (Receptor activator of nuclear factor kappa-B) inhibitor, a RANKL
(Receptor
activator of nuclear factor kappa-B ligand) inhibitor, an ERK inhibitor, a MAP
Kinase
inhibitor, an AKT inhibitor, a MEK inhibitor, a PI3K inhibitor, a HER1
inhibitor, a HER2
inhibitor, a HER3 inhibitor, a HER4 inhibitor, a Bc12 inhibitor, a CD22
inhibitor, a CD79b
inhibitor, an ErbB2 inhibitor, or a famesyl protein transferase inhibitor.
-68-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with any one or more of: 13-cis-retinoic acid, 345-
(methylsulfonylpiperadinemethyl)-indoly1]-quinolone, 4-hydroxytamoxifen, 5-
deooxyuridine, 5'-deoxy-5-fluorouridine, 5-fluorouracil, 6-mecaptopurine, 7-
hydroxystaurosporine, A-443654, abirateroneacetate, abraxane. ABT-578,
acolbifene, ADS-
100380, ALT-110, altretamine, amifostine, aminoglutethimide, amrubicin,
Amsacrine,
anagrelide, anastrozole, angiostatin, AP-23573, ARQ-197, arzoxifene, AS-
252424, AS-
605240, asparaginase, AT-9263, atrasentan, axitinib, AZD1152, Bacillus
Calmette-Guerin
(BCG) vaccine, batabulin, BC-210, besodutox, bevacizumab, bicalutamide, Bio 1
ii, BIO140,
bleomycin, BMS-214662, BMS-247550, BMS-275291, BMS-310705, bortezimib,
buserelin,
busulfan, calcitriol, camptothecin, canertinib, capecitabine, carboplatin,
carmustine, CC8490,
Cediranib, CG-1521, CG-781, chlamydocin, chlorambucil, chlorotoxin,
cilengitide,
cimitidine, cisplatin, cladribine, clodronate, COL-3, CP-724714,
cyclophosphamide,
cyproterone, cyproteroneacetate, cytarabine, qtosinearabinoside, dacarbazine,
dacinostat,
dactinomycin, dalotuztunab, danusertib, dasatanib, daunorubicin, decatanib,
deguelin,
denileukin, deoxycoformycin, depsipeptide, diarylpropionitrile,
diethylstilbestrol, diftitox,
docetaxel, dovitinib, doxorubicin, droloxifene, edotecarin, yttrium-90 labeled-
edotreotide,
edotreotide, EKB-569, EMD121974, endostatin, enzalutamide, enzastaurin,
epirubicin,
epithilone B, ERA-923, Erbitux, erlotinib, estradiol, estramustine, etoposide,
everolimus,
exemestane, ficlatuztunab, finasteride, flavopiridol, floxuridine,
fludarabine, fludrocortisone,
fluovmesterone, flutamide, FOLFOX regimen, Fulvestrant, galeterone, gefitinib,

gemcitabine, gimatecan, goserelin, goserelin acetate, gossypol, GSK461364,
GSK690693,
HMR-3339, hydroxyprogesteronecaproate, hydroxyurea, 187114, idarubicin,
idoxyfene,
ifosfamide, IM862, imatinib, IMC-1C11, INCB24360, IN01001, interferon,
interleukin-12,
ipilimtunab, irinotecan, JNJ-16241199, ketoconazole, KRX-0402, lapatinib,
lasofoxifene,
letrozole,leucovorin, leuprolide, leuprolide acetate, levamisole, liposome
entrapped
paclitaxel, lomustine, lonafarnib, lucanthone, LY292223, LY292696, LY293646,
LY293684,
LY294002, LY317615, marimastat, mechlorethamine, medroxyprogesteroneacetate,
megestrolacetate, melphalan, mercaptopurine, mesna, methotrexate, mithramycin,
mitomycin, mitotane, mitoxantrone, tozasertib, MLN8054, neovastat, Neratinib
neuradiab,
nilotinib, nilutimide, nolatrexed, NVP-BEZ235, oblimersen, octreotide,
ofattuntunab,
oregovomab, orteronel, oxaliplatin, paclitaxel, palbociclib, pamidronate,
panitumumab,
paionanib. PD0325901, PD184352, PEG-interferon, pemetrexed, pentostatin,
perifosine,
-69-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
phenylalaninemustard, PI-103, pictilisib, PIK-75, pipendoxifene, PKI-166,
plicamycin,
porfimer, prednisone, procarbazine, progesfins, PX-866, R-763, raloxifene,
ralfitrexed,
razoxin, ridaforolimus, rituximab, romidepsin, RTA744, rubitecan, scriptaid,
Sdx102,
seliciclib, selumetinib, semaxanib, SF1126, sirolimus, SN36093, sorafenib,
spironolactone,
squalamine, SR13668, streptozocin, SU6668, suberoylanalide hydroxamic acid,
sunitinib,
synthetic estrogen, talampanel, talimogene laherparepvec, tamoxifen,
temozolomide,
temsirolimus, teniposide, tesmilifene, testosterone, tetrandrine, TGX-221,
thalidomide,
thioguanine, thiotepa, ticilimumab, tipifarnib, tivozanib, TKI-258, TLK286,
topotecan,
toremifene citrate, trabectedin, trastuzumab, trefinoin, trichostatin A,
triciribinephosphate
monohydrate, triptorelin pamoate, TSE-424, uracil mustard, valproic acid,
valrubicin,
vandetanib, vatalanib, VEGF trap. vinblastine, vincristine, vindesine,
vinorelbine, vitaxin,
vitespan, vorinostat, VX-745, wortmannin, Xr311, zanolimumab, ZK186619, ZK-
304709,
ZM336372, ZSTK474.
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g., 14D7, 26B10 or humanized versions
thereof) is in
association with one or more antiemetics including, but not limited to:
casopitant
(GlaxoSmithKline), Netupitant (MGI-Helsinn) and other NK-1 receptor
antagonists,
palonosetron (sold as Aloxi by MGI Pharma), aprepitant (sold as Emend by Merck
and Co.;
Rahway, NJ), diphenhydramine (sold as Benadiy10 by Pfizer; New York, NY),
hydroxyzine
(sold as Atarax by Pfizer; New York, NY), metoclopramide (sold as Regime by
AH
Robins Co,; Richmond, VA), lorazepam (sold as Ativan by Wyeth; Madison, NJ),
alprazolam (sold as Xanax by Pfizer; New York, NY), haloperidol (sold as
Haldol by
Ortho-McNeil; Raritan, NJ), droperidol (Inapsinet), dronabinol (sold as
Marinol by Solvay
Pharmaceuticals, Inc.; Marietta, GA), dexamethasone (sold as Decadron by
Merck and Co.;
Rahway, NT), methylprednisolone (sold as Medrol by Pfizer; New York, NY),
prochlorperazine (sold as Compazine by Gla.xosinithkline; Research Triangle
Park, NC),
granisetron (sold as Kytril by Hoffmann-La Roche Inc.; Nutley, NJ),
ondansetron ( sold as
Zofrarie by Glaxosmithkline; Research Triangle Park, NC), dolasetron (sold as
Anzemet
by Sanofi-Aventis; New York, NY), tropisetron (sold as Navoban by Novartis;
East
Hanover, NJ).
Other side effects of cancer treatment include red and white blood cell
deficiency.
Accordingly, in an embodiment of the invention, an anti-TIGIT antibody or
antigen-binding
fragment thereof (e.g., 14D7, 26B10 or humanized versions thereof) is in
association with an
-70-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
agent which treats or prevents such a deficiency, such as, e.g., filgrastim,
PEG-filgrastim,
elythropoletin, epoetin alfa or darbepoetin alfa.
In an embodiment of the invention, an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g, 14D7, 26B10 or humanized versions
thereof) is
administered in association with anti-cancer radiation therapy. For example,
in an
embodiment of the invention, the radiation therapy is external beam therapy
(EBT): a method
for delivering a beam of high-energy X-rays to the location of the tumor. The
beam is
generated outside the patient (e.g., by a linear accelerator) and is targeted
at the tumor site.
These X-rays can destroy the cancer cells and careful treatment planning
allows the
surrounding normal tissues to be spared. No radioactive sources are placed
inside the
patient's body. In an embodiment of the invention, the radiation therapy is
proton beam
therapy: a type of conformal therapy that bombards the diseased tissue with
protons instead
of X-rays. In an embodiment of the invention, the radiation therapy is
conformal external
beam radiation therapy: a procedure that uses advanced technology to tailor
the radiation
therapy to an individual's body structures. In an embodiment of the invention,
the radiation
therapy is brachytherapy: the temporary placement of radioactive materials
within the body,
usually employed to give an extra dose¨or boost¨of radiation to an area.
In an embodiment of the invention, a surgical procedure administered in
association
with an anti-TIGIT antibody or antigen-binding fragment thereof (e.g., 14D7,
26B10 or
humanized versions thereof) is surgical tumorectomy.
The term "in association with" indicates that the components administered in a
method of the present invention (e.g., an anti-TIGIT antibody (e.g., humanized
antibody) or
antigen-binding fragment thereof (e.g., 14D7, 26B10 or humanized versions
thereof) along
with pembrolizumab) can be formulated into a single composition for
simultaneous delivery
or formulated separately into two or more compositions (e.g, a kit). Each
component can be
administered to a subject at a different time than when the other component is
administered;
for example, each administration may be given non-simultaneously (e.g.,
separately or
sequentially) at several intervals over a given period of time. Moreover, the
separate
components may be administered to a subject by the same or by a different
route.
Experimental and Diagnostic Uses
The anti-TIGIT antibodies and antigen-binding fragments thereof disclosed
herein
(e.g., 14D7, 26B10 and humanized versions thereof) may be used as affinity
purification
agents. in this process, the anti-TIGIT antibodies and antigen-binding
fragments thereof are
-71-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
immobilized on a solid phase such a Sephadex, glass or agarose resin or filter
paper, using
methods well known in the art. The immobilized antibody or fragment is
contacted with a
sample containing the TIGIT protein (or a fragment thereof) to be purified,
and thereafter the
support is washed with a suitable solvent that will remove substantially all
the material in the
sample except the TIGIT protein, which is bound to the immobilized antibody or
fragment.
Finally, the support is washed with a solvent which elutes the bound -MIT
(e.g., protein A).
Such immobilized antibodies and fragments form part of the present invention.
Further provided are antigens for generating secondary antibodies which are
useful
for example for performing Western blots and other immunoassays discussed
herein. In
particular, polypeptides are disclosed which comprise the variable regions
and/or CDR
sequences of a therapeutic antibody disclosed herein (e.g, 14D7, 26B10) and
which may be
used to generate anti-idiotypic antibodies for use in specifically detecting
the presence of the
antibody, e.g., in a therapeutic context.
Anti-TIGIT antibodies (e.g, humanized antibodies) and antigen-binding
fragments
thereof may also be useful in diagnostic assays for TIGIT protein, e.g.,
detecting its
expression in specific cells, tissues, or serum, e.g., tumor cells such as
melanoma cells. Such
diagnostic methods may be useful in various disease diagnoses.
The present invention includes ELISA assays (enzyme-linked immunosorbent
assay)
incorporating the use of an anti-TIGIT antibody or antigen-binding fragment
thereof
disclosed herein (e.g., 14D7, 26B10 or a humanized version thereof).
For example, such a method comprises the following steps:
(a) coat a substrate (e.g., surface of a microtiter plate well, e.g., a
plastic plate) with anti-
TIGIT antibody or antigen-binding fragment thereof:
(b) apply a sample to be tested for the presence of TIGIT to the substrate;
(c) wash the plate, so that unbound material in the sample is removed;
(d) apply detectably labeled antibodies (e.g., enzyme-linked antibodies) which
are also
specific to the MIT antigen;
(e) wash the substrate, so that the unbound, labeled antibodies are removed;
(1) if the labeled antibodies are enzyme linked, apply a chemical which is
converted by the
enzyme into a fluorescent signal; and
(g) detect the presence of the labeled antibody.
Detection of the label associated with the substrate indicates the presence of
the
TIGIT protein.
-72-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
In a further embodiment, the labeled antibody or antigen-binding fragment
thereof is
labeled with peroxidase which react with ABTS (e.g., 2,2'-azino-bis(3-
ethylbenzthiazoline-6-
sulphonic acid)) or 3,3',5,5'-Tetramethylbenzidine to produce a color change
which is
detectable. Alternatively, the labeled antibody or fragment is labeled with a
detectable
radioisotope (e.g., 3H) which can be detected by scintillation counter in the
presence of a
scintillant.
An anti-TIGIT antibody or antigen-binding fragment thereof of the invention
(e.g.,
14D7, 26B10 or humanized versions thereof) may be used in a Western blot or
immune-
protein blot procedure. Such a procedure forms part of the present invention
and includes
e.g.,:
(1) optionally transferring proteins from a sample to be tested for the
presence of TIGIT (e.g,
from a PAGE or SDS-PAGE electrophoretic separation of the proteins in the
sample) onto a
membrane or other solid substrate using a method known in the art (e.g., semi-
dry blotting or
tank blotting); contacting the membrane or other solid substrate to be tested
for the presence
of bound TIGIT or a fragment thereof with an anti-TIGIT antibody or antigen-
binding
fragment thereof of the invention.
Such a membrane may take the form of a nitrocellulose or vinyl-based (e.g.,
polyvinylidene fluoride (PVDF)) membrane to which the proteins to be tested
for the
presence of TIGIT in a non-denaturing PAGE (polyaciylamide gel
electrophoresis) gel or
SDS-PAGE (sodium dodecyl sulfate polyaaylamide gel electrophoresis) gel have
been
transferred (e.g., following electrophoretic separation in the gel). Before
contacting the
membrane with the anti-'TIGIT antibody or fragment, the membrane is optionally
blocked,
e.g., with non-fat dry milk or the like so as to bind non-specific protein
binding sites on the
membrane.
(2) washing the membrane one or more times to remove unbound anti-TIGIT
antibody or
fragment and other unbound substances; and
(3) detecting the bound anti-TIGIT antibody or fragment.
Detection of the bound antibody or fragment indicates that the TIGIT protein
is
present on the membrane or substrate and in the sample. Detection of the bound
antibody or
fragment may be by binding the antibody or fragment with a secondary antibody
(an anti-
immunoglobulin antibody) which is detectably labeled and, then, detecting the
presence of
the secondary antibody.
The anti-TIGIT antibodies and antigen-binding fragments thereof disclosed
herein
(e.g., 14D7, 26B10 and humanized versions thereof) may also be used for
-73-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
immunohistochemistiy. Such a method forms part of the present invention and
comprises,
e.g.,
(1) contacting a cell (e.g., a tumor cell such as a melanoma cell) to be
tested for the presence
of TIGIT protein with an anti-TIGIT antibody or antigen-binding fragment
thereof of the
invention; and
(2) detecting the antibody or fragment on or in the cell.
If the antibody or fragment itself is detectably labeled, it can be detected
directly.
Alternatively, the antibody or fragment may be bound by a detectably labeled
secondary
antibody which is detected.
Certain anti-TIGIT antibodies and antigen-binding fragments thereof disclosed
herein
(e.g., 14137, 26B10 and humanized versions thereof) may also be used for in
vivo tumor
imaging. Such a method may include injection of a radiolabeled anti-TIGIT
antibody or
antigen-binding fragment thereof into the body of a patient to be tested for
the presence of a
tumor associated with TIGIT expression (e.g., which expresses TIGIT, for
example, on the
tumor cell surface) followed by nuclear imaging of the body of the patient to
detect the
presence of the labeled antibody or fragment e.g., at loci comprising a high
concentration of
the antibody or fragment which are bound to the tumor. The detection of the
loci indicates
the presence of the TIGIT+ tumor and tumor cells.
Imaging techniques include SPECT imaging (single photon emission computed
tomography) or PET imaging (positron emission tomography). Labels include
e.g., iodine-
123 (1231) and technetium-99m (99mTc), e.g., in conjunction with SPECT imaging
or 11C, 13N,
150 or 18F, e.g., in conjunction with PET imaging or Indium-111 (See e.g.,
Gordon et al.,
(2005) International Rev. Neurobiol. 67:385-440).
Pharmaceutical Compositions and Administration
To prepare pharmaceutical or sterile compositions of the anti-TIGIT antibodies
and
antigen-binding fragments of the invention (e.g., 14D7, 26B10 and humanized
versions
thereof), the antibody or antigen-binding fragment thereof is admixed with a
pharmaceutically acceptable carrier or excipient. See, e.g., Remington's
Pharmaceutical
Sciences and US. Pharmacopeia: National Formulary, Mack Publishing Company,
Easton,
PA (1984).
Formulations of therapeutic and diagnostic agents may be prepared by mixing
with
acceptable carriers, excipients, or stabilizers in the form of, e.g.,
lyophilized powders,
slurries, aqueous solutions or suspensions (see, e.g., Hardman, etal. (2001)
Goodman and
-74-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Gilman 's The Pharmacological Basis of Therapeutics, McGraw-Hill, New York,
NY;
Gennaro (2000) Remington: The Science and Practice of Pharmacy, Lippincott,
Williams,
and Wilkins, New York, NY; Avis, etal. (eds.) (1993) Pharmaceutical Dosage
Forms:
Parenteral Medications, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990)
Pharmaceutical
Dosage Forms: Tablets, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990)
Pharmaceutical
Dosage Forms: Disperse Systems, Marcel Dekker, NY; Weiner and Kotkoskie (2000)
Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, NY).
Toxicity and therapeutic efficacy of the antibodies of the invention,
administered
alone or in combination with another therapeutic agent, can be determined by
standard
pharmaceutical procedures in cell cultures or experimental animals, e.g, for
determining the
LD50 (the dose lethal to 50% of the population) and the ED50 (the dose
therapeutically
effective in 50% of the population). The dose ratio between toxic and
therapeutic effects is
the therapeutic index (LD50/ ED50). The data obtained from these cell culture
assays and
animal studies can be used in formulating a range of dosage for use in human.
The dosage of
such compounds lies preferably within a range of circulating concentrations
that include the
ED50 with little or no toxicity. The dosage may vary within this range
depending upon the
dosage form employed and the route of administration.
In a further embodiment, a further therapeutic agent that is administered to a
subject
in association with an anti-TIGIT antibody or antigen-binding fragment thereof
of the
invention (e.g., 14D7, 261310) in accordance with the Physicians' Desk
Reference 2003
(Thomson Healthcare; 57th edition (November 1, 2002)).
The mode of administration can vary. Routes of administration include oral,
rectal,
transmucosal, intestinal, parenteral; intramuscular, subcutaneous,
intradermal, intramedullaty,
intrathecal, direct intraventricular, intravenous, intraperitoneal,
intranasal, intraocular,
inhalation, insufflation, topical, cutaneous, transdermal, or intra-arterial.
In particular embodiments, the anti-TIGIT antibodies or antigen-binding
fragments
thereof of the invention (e.g., 14D7, 261310 and humanized versions thereof)
can be
administered by an invasive route such as by injection. In further embodiments
of the
invention, an anti- TIGIT antibody or antigen-binding fragment thereof, or
pharmaceutical
composition thereof, is administered intravenously, subcutaneously,
intramuscularly,
intraarterially, intratumorally, or by inhalation, aerosol delively.
Administration by non-
invasive routes (e.g., orally; for example, in a pill, capsule or tablet) is
also within the scope
of the present invention.
-75-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
The present invention provides a vessel (e.g., a plastic or glass vial, e.g.,
with a cap or
a chromatography column, hollow bore needle or a syringe cylinder) comprising
any of the
antibodies or antigen-binding fragments of the invention (e.g., 14D7, 26B10
and humanized
versions thereof) or a pharmaceutical composition thereof. The present
invention also
provides an injection device comprising any of the antibodies or antigen-
binding fragments of
the invention (e.g., 14D7, 26B10 and humanized versions thereof) or a
pharmaceutical
composition thereof. An injection device is a device that introduces a
substance into the body
of a patient via a parenteral route, e.g., intramuscular, subcutaneous or
intravenous. For
example, an injection device may be a syringe (e.g., pre-filled with the
pharmaceutical
composition, such as an auto-injector) which, for example, includes a cylinder
or barrel for
holding fluid to be injected (e.g, antibody or fragment or a pharmaceutical
composition
thereof), a needle for piecing skin and/or blood vessels for injection of the
fluid; and a
plunger for pushing the fluid out of the cylinder and through the needle bore.
In an
embodiment of the invention, an injection device that comprises an antibody or
antigen-
binding fragment thereof of the present invention or a pharmaceutical
composition thereof is
an intravenous (IV) injection device. Such a device includes the antibody or
fragment or a
pharmaceutical composition thereof in a cannula or trocar/needle which may be
attached to a
tube which may be attached to a bag or reservoir for holding fluid (e.g,
saline; or lactated
ringer solution comprising NaCl, sodium lactate, KCI, CaCl2 and optionally
including
glucose) introduced into the body of the patient through the cannula or
trocar/needle. The
antibody or fragment or a pharmaceutical composition thereof may, in an
embodiment of the
invention, be introduced into the device once the trocar and cannula are
inserted into the vein
of a subject and the trocar is removed from the inserted cannula. The IV
device may, for
example, be inserted into a peripheral vein (e.g, in the hand or arm); the
superior vena cava
or inferior vena cava, or within the right atrium of the heart (e.g., a
central IV); or into a
subclavian, internal jugular, or a femoral vein and, for example, advanced
toward the heart
until it reaches the superior vena cava or right atrium (e.g., a central
venous line). In an
embodiment of the invention, an injection device is an autoinjector; a jet
injector or an
external infusion pump. A jet injector uses a high-pressure narrow jet of
liquid which
penetrate the epidermis to introduce the antibody or fragment or a
pharmaceutical
composition thereof to a patient's body. External infusion pumps are medical
devices that
deliver the antibody or fragment or a pharmaceutical composition thereof into
a patient's
body in controlled amounts. External infusion pumps may be powered
electrically or
mechanically. Different pumps operate in different ways, for example, a
syringe pump holds
-76-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
fluid in the reservoir of a syringe, and a moveable piston controls fluid
delivery, an
elastomeric pump holds fluid in a stretchable balloon reservoir, and pressure
from the elastic
walls of the balloon drives fluid delivery. In a peristaltic pump, a set of
rollers pinches down
on a length of flexible tubing, pushing fluid forward. In a multi-channel
pump, fluids can be
delivered from multiple reservoirs at multiple rates.
The pharmaceutical compositions disclosed herein may also be administered with
a
needleless hypodermic injection device; such as the devices disclosed in U.S.
Patent Nos.
6,620,135; 6,096,002; 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880;
4,790,824 or
4,596,556. Such needleless devices comprising the pharmaceutical composition
are also part
of the present invention. The
pharmaceutical compositions disclosed herein may also be
administered by infusion. Examples of well-known implants and modules for
administering
the pharmaceutical compositions include those disclosed in: U.S. Patent No.
4,487,603,
which discloses an implantable micro-infusion pump for dispensing medication
at a
controlled rate; U.S. Patent No. 4,447,233, which discloses a medication
infusion pump for
delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224,
which discloses a
variable flow implantable infusion apparatus for continuous drug delivery;
U.S. Patent. No.
4,439,196, which discloses an osmotic drug delivery system having multi-
chamber
compartments. Many other such implants, delivery systems, and modules are well
known to
those skilled in the art and those comprising the pharmaceutical compositions
of the present
invention are within the scope of the present invention.
Alternately, one may administer the anti-TIGIT antibody or antigen-binding
fragment
of the invention (e.g., 14D7, 26B10 or humanized versions thereof) in a local
rather than
systemic manner, for example, via injection of the antibody or fragment
directly into a tumor,
e.g., a TIGIT+ tumor. Furthermore, one may administer the antibody or fragment
in a
targeted drug delivery system, for example, in a liposome coated with a tissue-
specific
antibody, targeting, for example, a tumor e.g., a TIGIT+ tumor, e.g.,
characterized by
immunopathology. The liposomes will be targeted to and taken up selectively by
the
afflicted tissue. Such methods and liposomes are part of the present
invention.
The administration regimen depends on several factors, including the serum or
tissue
turnover rate of the therapeutic antibody or antigen-binding fragment, the
level of symptoms,
the inununogenicity of the therapeutic antibody, and the accessibility of the
target cells in the
biological matrix. Preferably, the administration regimen delivers sufficient
therapeutic
antibody or fragment to effect improvement in the target disease state, while
simultaneously
minimizing undesired side effects. Accordingly, the amount of biologic
delivered depends in
-77-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
part on the particular therapeutic antibody and the severity of the condition
being treated.
Guidance in selecting appropriate doses of therapeutic antibodies or fragments
is available
(see, e.g., Wawzynczak (1996)Antibody Therapy, Bios Scientific Pub. Ltd,
Oxfordshire,
UK; Kresina (ed.) (1991) Monoclonal Antibodies, Cytokines and Arthritis,
Marcel Dekker,
New York, NY; Bach (ed.) (1993)Monoclonal Antibodies and Peptide Therapy in
Autoimmune Diseases, Marcel Dekker, New York, NY; Baert, et al. (2003) New
Engl. J.
Med. 348:601-608; Milgrom etal. (1999) New Engl. J. Med. 341:1966-1973; Slamon
etal.
(2001) New Engl. J. Med. 344:783-792; Beniaminovitz etal. (2000) New Engl. J.
Med.
342:613-619; Ghosh etal. (2003) New Engl. J. Med. 348:24-32; Lipsky et al.
(2000) New
Engl. J Med. 343:1594-1602).
Determination of the appropriate dose is made by the clinician, e.g, using
parameters
or factors known or suspected in the art to affect treatment. Generally, the
dose begins with
an amount somewhat less than the optimum dose and it is increased by small
increments
thereafter until the desired or optimum effect is achieved relative to any
negative side effects.
Important diagnostic measures include those of symptoms of, e.g., the
inflammation or level
of inflammatory cytokines produced. In general, it is desirable that a
biologic that will be
used is derived from the same species as the animal targeted for treatment,
thereby
minimizing any immune response to the reagent. In the case of human subjects,
for example,
humanized and fully human antibodies are may be desirable.
Antibodies or antigen-binding fragments thereof disclosed herein (e.g., 14D7,
26B10
and humanized versions thereof) may be provided by continuous infusion, or by
doses
administered, e.g., daily, 1-7 times per week, weekly, bi-weekly, monthly,
bimonthly,
quarterly, semiannually, annually etc. Doses may be provided, e.g.,
intravenously,
subcutaneously, topically, orally, nasally, rectally, intramuscular,
intracerebrally,
intraspinally, or by inhalation. A total weekly dose is generally at least
0.05 jig/kg body
weight, more generally at least 0.2 jig/kg, 0.5 jig/kg, 1 jig/kg, 10 jig/kg,
100 jig/kg, 0.25
mg/kg, 1.0 mg/kg, 2.0 mg/kg, 5.0 mg/ml, 10 mg/kg, 25 mg/kg, 50 mg/kg or more
(see, e.g.,
Yang, et al. (2003) New Engl. J. Med 349:427-434; Herold, et al. (2002) New
Engl. J. Med.
346:1692-1698; Liu, et al. (1999)J. Neurol. Neurosurg. Psych. 67:451-456;
Portielji, etal.
(20003) Cancer Immunol. Immunother. 52:151-144). Doses may also be provided to
achieve
a pre-determined target concentration of anti-TIGIT antibody in the subject's
serum, such as
0.1, 0.3, 1, 3, 10, 30, 100, 300 jig/ml or more. In other embodiments, An anti-
T1GIT
antibody of the present invention is administered, e.g., subcutaneously or
intravenously, on a
-78-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
weekly, biweekly, "every 4 weeks," monthly, bimonthly, or quarterly basis at
10, 20, 50, 80,
100, 200, 500, 1000 or 2500 mg/subject.
As used herein, the term "effective amount" refer to an amount of an anti-
TIGIT or
antigen-binding fragment thereof of the invention (e.g., humanized 14D7 or
humanized
26B10) that, when administered alone or in combination with an additional
therapeutic agent
to a cell, tissue, or subject, is effective to cause a measurable improvement
in one or more
symptoms of disease, for example cancer or the progression of cancer. An
effective dose
further refers to that amount of the antibody or fragment sufficient to result
in at least partial
amelioration of symptoms, e.g., tumor shrinkage or elimination, lack of tumor
growth,
increased survival time. When applied to an individual active ingredient
administered alone,
an effective dose refers to that ingredient alone. When applied to a
combination, an effective
dose refers to combined amounts of the active ingredients that result in the
therapeutic effect,
whether administered in combination, serially or simultaneously. An effective
amount of a
therapeutic will result in an improvement of a diagnostic measure or parameter
by at least
10%; usually by at least 20%; preferably at least about 30%; more preferably
at least 40%,
and most preferably by at least 50%. An effective amount can also result in an
improvement
in a subjective measure in cases where subjective measures are used to assess
disease
severity.
Kits
Further provided are kits comprising one or more components that include, but
are not
limited to, an anti-TIGIT antibody or antigen-binding fragment, as discussed
herein (e.g.,
humanized 14D7 or humanized 26B10) in association with one or more additional
components including, but not limited to a pharmaceutically acceptable carrier
and/or a
therapeutic agent, as discussed herein. The antibody or fragment and/or the
therapeutic agent
can be formulated as a pure composition or in combination with a
pharmaceutically
acceptable carrier, in a pharmaceutical composition.
In one embodiment, the kit includes an anti-TIGIT antibody or antigen-binding
fragment thereof of the invention (e.g., humanized 14D7 or humanized 26B10) or
a
pharmaceutical composition thereof in one container (e.g., in a sterile glass
or plastic vial)
and a pharmaceutical composition thereof and/or a therapeutic agent in another
container
(e.g., in a sterile glass or plastic vial).
In another embodiment, the kit comprises a combination of the invention,
including
an anti-TIGIT antibody or antigen-binding fragment thereof of the invention
(e.g., humanited
-79-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
14D7 or humanized 26B10) along with a pharmaceutically acceptable carrier,
optionally in
combination with one or more therapeutic agents formulated together,
optionally, in a
pharmaceutical composition, in a single, common container.
If the kit includes a pharmaceutical composition for parenteral administration
to a
subject, the kit can include a device for performing such administration. For
example, the kit
can include one or more hypodermic needles or other injection devices as
discussed above.
The kit can include a package insert including information concerning the
pharmaceutical compositions and dosage forms in the kit. Generally, such
information aids
patients and physicians in using the enclosed pharmaceutical compositions and
dosage forms
effectively and safely. For example, the following information regarding a
combination of
the invention may be supplied in the insert: pharmacokinetics,
pharmacodynamics, clinical
studies, efficacy parameters, indications and usage, contraindications,
warnings, precautions,
adverse reactions, overdosage, proper dosage and administration, how supplied,
proper
storage conditions, references, manufacturer/distributor information and
patent information.
Detection Kits and Therapeutic Kits
As a matter of convenience, an anti-TIGIT antibody or antigen-binding fragment

thereof of the invention (e.g., 14D7, 26B10 or humanized versions thereof) can
be provided
in a kit, i.e., a packaged combination of reagents in predetermined amounts
with instructions
for performing the diagnostic or detection assay. Where the antibody or
fragment is labeled
with an enzyme, the kit will include substrates and cofactors required by the
enzyme (e.g., a
substrate precursor which provides the detectable chromophore or fluorophore).
In addition,
other additives may be included such as stabilizers, buffers (e.g., a block
buffer or lysis
buffer) and the like. The relative amounts of the various reagents may be
varied widely to
provide for concentrations in solution of the reagents which substantially
optimize the
sensitivity of the assay. Particularly, the reagents may be provided as dry
powders, usually
lyophilized, including excipients which on dissolution will provide a reagent
solution having
the appropriate concentration.
Also provided are diagnostic or detection reagents and kits comprising one or
more
such reagents for use in a variety of detection assays, including for example,
immunoassays
such as ELISA (sandwich-type or competitive format). The kit's components may
be pre-
attached to a solid support, or may be applied to the surface of a solid
support when the kit is
used. In some embodiments of the invention, the signal generating means may
come pre-
associated with an antibody or fragment of the invention or may require
combination with
-80-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
one or more components, e.g., buffers, antibody-enzyme conjugates, enzyme
substrates, or
the like, prior to use. Kits may also include additional reagents, e.g.,
blocking reagents for
reducing nonspecific binding to the solid phase surface, washing reagents,
enzyme substrates,
and the like. The solid phase surface may be in the form of a tube. a bead, a
microtiter plate,
a microsphere, or other materials suitable for immobilizing proteins,
peptides, or
3.0 polypeptides. In particular aspects, an enzyme that catalyzes the
formation of a
chemilluminescent or chromogenic product or the reduction of a
chemilltuninescent or
chromogenic substrate is a component of the signal generating means. Such
enzymes are well
known in the art. Kits may comprise any of the capture agents and detection
reagents
described herein. Optionally the kit may also comprise instructions for
carrying out the
methods of the invention.
Also provided is a kit comprising an anti-TIGIT antibody (e.g., humanized
antibody)
or antigen-binding fragment thereof packaged in a container, such as a vial or
bottle, and
further comprising a label attached to or packaged with the container, the
label describing the
contents of the container and providing indications and/or instructions
regarding use of the
contents of the container to treat one or more disease states as described
herein.
In one aspect, the kit is for treating cancer and comprises an anti-TIGIT
antibody
(e.g., humanized antibody) or antigen-binding fragment thereof and a further
therapeutic
agent or a vaccine. The kit may optionally further include a syringe for
parenteral, e.g.,
intravenous, administration. In another aspect, the kit comprises an anti-
TIGTT antibody
(e.g, humanized antibody) or antigen-binding fragment thereof and a label
attached to or
packaged with the container describing use of the antibody or fragment with
the vaccine or
further therapeutic agent. In yet another aspect, the kit comprises the
vaccine or further
therapeutic agent and a label attached to or packaged with the container
describing use of the
vaccine or further therapeutic agent with the anti-TIG1T antibody or fragment.
In certain
embodiments, an anti-TIGIT antibody and vaccine or further therapeutic agent
are in separate
vials or are combined together in the same pharmaceutical composition.
As discussed above in the combination therapy section, concurrent
administration of
two therapeutic agents does not require that the agents be administered at the
same time or by
the same route, as long as there is an overlap in the time period during which
the agents are
exerting their therapeutic effect. Simultaneous or sequential administration
is contemplated,
as is administration on different days or weeks.
The therapeutic and detection kits disclosed herein may also be prepared that
comprise at least one of the antibody, peptide, antigen-binding fragment, or
polynucleotide
-81-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
disclosed herein and instructions for using the composition as a detection
reagent or
therapeutic agent. Containers for use in such kits may typically comprise at
least one vial, test
tube, flask, bottle, syringe or other suitable container, into which one or
more of the detection
and/or therapeutic composition(s) may be placed, and preferably suitably
aliquoted. Where a
second therapeutic agent is also provided, the kit may also contain a second
distinct container
into which this second detection and/or therapeutic composition may be placed.
Alternatively, a plurality of compounds may be prepared in a single
pharmaceutical
composition, and may be packaged in a single container means, such as a vial,
flask, syringe,
bottle, or other suitable single container. The kits disclosed herein will
also typically include
a means for containing the vial(s) in close confinement for commercial sale,
such as, e.g.,
injection or blow-molded plastic containers into which the desired vial(s) are
retained. Where
a radiolabel, chromogenic, fluorigenic, or other type of detectable label or
detecting means is
included within the kit, the labeling agent may be provided either in the same
container as the
detection or therapeutic composition itself, or may alternatively be placed in
a second distinct
container means into which this second composition may be placed and suitably
aliquoted.
Alternatively, the detection reagent and the label may be prepared in a single
container
means, and in most cases, the kit will also typically include a means for
containing the vial(s)
in close confinement for commercial sale and/or convenient packaging and
delivery.
A device or apparatus for carrying out the detection or monitoring methods
described
herein is also provided. Such an apparatus may include a chamber or tube into
which sample
can be input, a fluid handling system optionally including valves or pumps to
direct flow of
the sample through the device, optionally filters to separate plasma or serum
from blood,
mixing chambers for the addition of capture agents or detection reagents, and
optionally a
detection device for detecting the amount of detectable label bound to the
capture agent
irrununocomplex. The flow of sample may be passive (e.g, by capillary,
hydrostatic, or other
forces that do not require further manipulation of the device once sample is
applied) or active
(e.g., by application of force generated via mechanical pumps, electroosmotic
pumps,
centrifugal force, or increased air pressure), or by a combination of active
and passive forces.
In further embodiments, also provided is a processor, a computer readable
memory,
and a routine stored on the computer readable memory and adapted to be
executed on the
processor to perform any of the methods described herein. Examples of suitable
computing
systems, environments, and/or configurations include personal computers,
server computers,
hand-held or laptop devices, multiprocessor systems, microprocessor-based
systems, set top
boxes, programmable consumer electronics, network PCs, minicomputers,
mainframe
-82-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
computers, distributed computing environments that include any of the above
systems or
devices, or any other systems known in the art.
GENERAL METHODS
Standard methods in molecular biology are described Sambrook, Fritsch and
Maniatis
(1982 & 1989 2nd Edition, 2001 3"tEdition) Molecular Cloning. A Laboratory
Manual, Cold
Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Sambrook and Russell
(2001)
Molecular Cloning, Yed ed., Cold Spring Harbor Laboratory Press, Cold Spring
Harbor, NY;
Wu (1993) Recombinant DNA, Vol. 217, Academic Press, San Diego, CA). Standard
methods also appear in Ausbel, et al. (2001) Current Protocols in Molecular
Biology, Vols.1-
4, John Wiley and Sons, Inc. New York, NY, which describes cloning in
bacterial cells and
DNA mutagenesis (Vol. 1), cloning in mammalian cells and yeast (Vol. 2),
glycoconjugates
and protein expression (Vol. 3), and bioinformatics (Vol. 4).
Methods for protein purification including inununoprecipitation;
chromatography,
electrophoresis, centrifugation, and crystallization are described (Coligan,
et al. (2000)
Current Protocols in Protein Science, Vol. 1, John Wiley and Sons, Inc., New
York).
Chemical analysis, chemical modification, post-translational modification,
production of
fusion proteins, glycosylation of proteins are described (see, e.g, Coligan,
etal. (2000)
Current Protocols in Protein Science, Vol. 2, John Wiley and Sons, Inc., New
York; Ausubel,
et al. (2001) Current Protocols in Molecular Biology, Vol. 3, John Wiley and
Sons, Inc., NY,
NY, pp. 16Ø5-16.22.17; Sigma-Aldrich, Co. (2001) Products for Life Science
Research, St.
Louis, MO; pp. 45-89; Amersham Pharmacia Biotech (2001) BioDirectory,
Piscataway, N.J.,
pp. 384-391). Production, purification, and fragmentation of polyclonal and
monoclonal
antibodies are described (Coligan, etal. (2001) Current Protcols in
Immunology. Vol. I, John
Wiley and Sons, Inc., New York; Harlow and Lane (1999) Using Antibodies, Cold
Spring
Harbor Laboratory Press, Cold Spring Harbor, NY; Harlow and Lane, supra).
Standard
techniques for characterizing ligandireceptor interactions are available (see,
e.g., Coligan, et
al. (2001) Current Protocols in Immunology, Vol. 4, John Wiley, Inc., New
York).
Monoclonal, polyclonal, and humanized antibodies can be prepared (see, e.g.,
Sheperd and Dean (eds.) (2000) Monoclonal Antibodies, Oxford Univ. Press, New
York, NY;
Kontennarm and Dubel (eds.) (2001) Antibody Engineering, Springer-Verlag, New
York;
Harlow and Lane (1988) Antibodies A Laboratory Manual, Cold Spring Harbor
Laboratory
Press, Cold Spring Harbor, NY, pp. 139-243; Carpenter, etal. (2000)J. Immunol.
165:6205;
He, etal. (1998) J. Immunol. 160:1029: Tang et al. (1999) J. Biol. Chem.
274:27371-27378;
-83-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Baca et at. (1997)J. Biol. Chem. 272:10678-10684; Chothia et at. (1989) Nature
342:877-
883; Foote and Winter (1992) J. Mot. Biol. 224:487-499; U.S. Pat. No.
6,329,511).
An alternative to humanization is to use human antibody libraries displayed on
phage
or human antibody libraries in transgenic mice (Vaughan et at. (1996) Nature
Biotechnol.
14:309-314; Barbas (1995) Nature Medicine 1:837-839; Mendez et at. (1997)
Nature
Genetics 15:146-156; Hoogenboom and Chames (2000) ImmunoL Today 21:371-377;
Barbas
et at. (2001) Phage Display A Laboratory Manual, Cold Spring Harbor Laboratory
Press,
Cold Spring Harbor, New York; Kay et at. (1996) Phage Display of Peptides and
Proteins: A
Laboratory Manual, Academic Press, San Diego, CA; de Bruin etal. (1999) Nature

BiotechnoL 17:397-399).
Single chain antibodies and diabodies are described (see, e.g., Malecki et at.
(2002)
Proc. Natl. Acad Sci. USA 99:213-218; Conrath et at. (2001)J. BioL Chem.
276:7346-7350;
Desmy ter etal. (2001)J. Biol. (hem. 276:26285-26290; Hudson and Kortt
(1999)J.
ImmunoL Methods 231:177-189; and U.S. Pat. No. 4,946,778). Bifunctional
antibodies are
provided (see, e.g., Mack, et at. (1995) Proc. NatL Acad. Sci. USA 92:7021-
7025; Carter
(2001)J. ImmunoL Methods 248:7-15; Volkel, etal. (2001) Protein Engineering
14:815-823;
Segal, et at. (2001)J. ImmunoL Methods 248:1-6; Brennan, et at. (1985) Science
229:81-83;
Raso, et at. (1997)J. Biol. Chem. 272:27623; Morrison (1985) Science 229:1202-
1207;
Traunecker, et at. (1991) EMBO J. 10:3655-3659; and U.S. Pat. Nos. 5,932,448,
5,532,210,
and 6,129,914).
Bispecific antibodies are also provided (see, e.g, Azzoni et at. (1998)J.
Immunol.
161:3493; Kita etal. (1999)J. ImmunoL 162:6901; Merchant et at. (2000)J. BioL
Chem.
74:9115; Pandey el at. (2000)J. Biol. Chem. 275:38633; Zheng el at. (2001)J.
Blot Chem.
276:12999; Propst et at. (2000)J Immunot 165:2214; Long (1999) Ann. Rev.
ImmunoL
17:875).
Purification of antigen is not necessary for the generation of antibodies.
Animals can
be immunized with cells bearing the antigen of interest. Splenocytes can then
be isolated
from the immunized animals, and the splenocytes can fused with a myeloma cell
line to
produce a hybridoma (see, e.g., Meyaard etal. (1997) Immunity 7:283-290;
Wright et at.
(2000) Immunity 13:233-242; Preston et at., supra; Kaithamana et at. (1999)J.
ImmunoL
163:5157-5164).
Antibodies can be conjugated, e.g, to small drug molecules, enzymes,
liposomes,
polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic,
kit or other
purposes, and include antibodies coupled, e.g., to dyes, radioisotopes,
enzymes, or metals,
-84-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
e.g., colloidal gold (see, e.g., Le Doussal et at (1991)1 Immunol. 146:169-
175; GibeRini et
al. (1998).1. Immunol. 160:3891-3898; Hsing and Bishop (1999)1 Immunol.
162:2804-2811;
Everts et al. (2002)1 Immunol. 168:883-889).
Methods for flow cytometry, including fluorescence activated cell sorting
(FACS), are
available (see, e.g., Owens, et al. (1994) Flow C'ytometry Principles for
Clinical Laboratory
Practice, John Wiley and Sons, Hoboken, NJ; Givan (2001) Flow (ytometry, 2"d
ed.; Wiley-
Liss, Hoboken, NJ; Shapiro (2003) Practical Flow Cytometry, john Wiley and
Sons,
Hoboken, NJ). Fluorescent reagents suitable for modifying nucleic acids,
including nucleic
acid primers and probes, polypeptides, and antibodies, for use, e.g., as
diagnostic reagents,
are available (Molecular Probes (2003) Catalogue, Molecular Probes, Inc.,
Eugene, OR;
Sigma-Aldrich (2003) Catalogue, St. Louis, MO).
Standard methods of histology of the immune system are described (see, e.g.,
Muller-
Harmelink (ed.) (1986) Human Thymus: Histopathology and Pathology, Springer
Verlag,
New York, NY; Hiatt, et al. (2000) Color Atlas of Histology, Lippincott,
Williams, and
Wilkins, Phila, PA; Louis, et al. (2002) Basic Histology: Text and Atlas,
McGraw-Hill, New
York, NY).
Software packages and databases for determining, e.g., antigenic fragments,
leader
sequences, protein folding, functional domains, glycosylation sites, and
sequence alignments,
are available (see, e.g., GenBank, Vector NTI Suite (Informax, Inc, Bethesda,
MD); GCG
Wisconsin Package (Accelrys, Inc., San Diego, CA); DeCypher (TimeLogic Corp.,
Crystal
Bay, Nevada); Menne, et al. (2000) Bioinformatics 16: 741-742; Menne, etal.
(2000)
Bioinformatics Applications Note 16:741-742; Wren, et al. (2002) Comput
Methods
Programs Biomed. 68:177-181; von Heijne (1983) Eur. .1. Biochem. 133:17-21;
von Heijne
(1986) Nucleic Acids Res. 14:4683-4690).
Example 1
Generation of Mouse anti-hTIGIT Antibodies
To generate antibodies to human TIGIT, Balb/C mice were immunized with human
Fc-tagged human TIGIT recombinant protein using RIBI adjuvant and footpad
injection on a
biweekly schedule. Immunized animals were bled and serum titers determined for
binding to
human TIGIT transfected CHOK1 cells using a cell-based ELISA (described
below). The
animals with the highest titers were given a final boost with recombinant
protein and draining
popliteal lymph nodes isolated four days later. Hybridomas were generated by
electrofusion
-85-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
of isolated lymphocytes with the myeloma fusion partner P3X63-AG8.653 using
the
Cytopulse Hybrimmune electrofusion system. Fused cells were plated in 96-well
plates in
DMEM/F12, 15% BCS, HAT, IL-6, OPI supplement, and gentamycin.
Hybridoma supernatants were assayed for binding to human TIGIT expressing
CHOK1 cells and cross-reactivity to cynomolgous and rhesus TIGIT expressing
CHO cells
using a cell-based ELISA format. Human -MIT and cyno/rhesus TIGIT expressing
CHO-
K1 cells were plated in 96-well tissue-culture plates in 50 [11 of DMEM/F12,
10% BCS and
gentamycin (CHO-Kl media). Cells were plated at either 2x104 cells/well two
days prior to
the assay or 4x104 cells/well one day prior to the assay. Media was removed
from the wells
prior to the assay and 501.d of hybridoma supernatant added. Hybridoma
supernatants were
incubated for 30-60 minutes at room temperature and washed 3 times with
PBS/0.05%
Tween 20 using a cell ELISA washing protocol on the Biotek EL405x Select CW
plate
washer. Fifty microliters of the detection antibody (HRP-conjugated goat anti-
rat IgG
(Southern Biotech cat# 3030-05) or HRP-conjugated goat anti-mouse IgG
(Southern Biotech
cat# 1043-05)), was added at a 1:2000 dilution in CHO-Kl media and incubated
at room
temperature for 30-60 minutes. Assay plates were washed as above and developed
with
TMB and stopped with TMB stop solution (KPL cat# 50-85-06) or 0.1N phosphoric
acid.
The absorbance at 450nm-620nm was determined. Positive clones were reactive to
both
human TIGIT and cyno/rhesus TIGIT transfected CHO-Kl cells, and were negative
for
binding to parental CHO-Kl cells. In these assays, if an antibody showed
binding to parental
(untransfected) CHO-Kl cells: we discarded that antibody in screening as not
specific to
TIGIT.
Positive hybridomas were subcloned by limiting dilution or subcloned by
plating
hybridomas in semi-solid media and clones picked on the ClonePix (Genetix).
Two rounds
of subcloning were performed on the parental hybridomas. Final subclones were
grown in
small-scale cultures in serum-free hybridoma production medium and purified to
generate
purified antibody for further characterization.
-86-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Example 2
Characterization of anti-hTIGIT Antibodies
Supematants from positive clones were tested for their ability to block
recombinant
human CD155-huFc protein binding to hTIGIT CHOK1 cells in a cell-based ELISA
format.
Human TIGIT-CHO-Kl cells were plated in 96-well plates as described above.
Media was
removed from the plates and 50 1.t1 of hybridoma supematant was incubated with
the human
TIGIT CHO-Kl cells at 4 C for 30 minutes. Fifty microliters of human CD155-
huFc was
added to the plate for a final concentration of 0.51.1g/m1 of human CD155-huFc
and incubated
for 30 minutes at 4 C. Assay plates were washed 3 times with PBS/0.05% Tween-
20 as
above. Binding of human CD155-huFc to the hTIGIT-CHOK1 cells was detected
using an
HRP-conjugate F(ab)"2 goat anti-human IgG secondary antibody (Jackson 109-036-
098) at
1:2000 dilution in CHO-K1 media. Plates were developed using TMB and stopped
using
TMB Stop Solution as desciived above and the A450-620nm determined.
A mouse antibody generated according to the above described method is referred
to as
14D7, and was derived from clone MEB125.14D7. This mouse antibody (14D7) is of
the
IgGl/kappa isotype and comprises the heavy chain variable region of SEQ ID
NO:6 and the
light chain variable region of SEQ ID NO:?. Purified 14D7 antibody binds to
human TIGIT
and cyno/rhesus TIGIT as determined by cell-based ELISA binding to human TIGIT
and
cyno/rhesus TIGIT-CHOK1 cells (Figure 1) using the methods described above.
Purified
14D7 antibody also block the hTIGIT and hCD155 interaction using a cell-based
ELISA
blocking assay (Figure 2) using the method described above.
Another mouse antibody generated according to the above described method is
referred to as 26B10, and was derived from clone MEB125.26B10. This mouse
antibody
(26B10) is of the IgGI/kappa isotype and comprises the heavy chain variable
region of SEQ
ID NO:23 and the light chain variable region of SEQ ID NO:24. Purified 26B10
antibody
binds to human TIGIT and cyno/rhesus TIGIT as determined by cell-based ELISA
binding to
human MIT and cyno/rhesus TIGIT-CHOK1 cells (Figure 1) using the methods
described
above. Purified 26B10 antibody also blocks the hTIGIT and hCD155 interaction
using a cell-
based ELISA blocking assay (Figure 2) using the method described above.
Affinity determination for binding of parental (non-human) anti-TIGIT
antibodies to human TWIT recombinant protein: The kinetic binding activity of
anti-
human TIGIT antibodies 14D7 and 26B10 (made as described in Example 1) and of
commercial antibody MBSA43 was measured by surface plasmon resonance using a
Biacore
-87-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
T200 system (Biacore, GE Healthcare, Piscataway, NJ). Approximately 5000 RU of
Anti-
mouse IgG, GE Healthcare Catalog Number BR-1008-38, or approximately 13,000 RU
of
Goat Anti-Rat IgG Fc gamma, Fragment Specific, Jackson ImmunoResearch Catalog
Number 112-006-071, was immobilized via amine coupling chemistry onto a Series
S CM5
sensor chip, catalog number BR-1005-30. Mouse anti-human TIGIT clones, 14D7,
26B10
and MBSA43, were each injected over the immobilized anti-mouse surfaces at 1
ttglmL for a
capture level of 40 RU. HBS-EP+ buffer (BR-1006-69) was used as the running
buffer with
a flow rate of 30A/min. Varying concentrations of human TIGIT-His protein,
ranging from
0.29 nM to 40 nM, at a flow rate of 45A/rain were injected over the antibody
surfaces.
Following each Mouse anti-human TIGIT injection cycle, the Series S CM5 chip
surface was
regenerated using one three- minute injection of 10mM Glycine pH 1.7 at a flow
rate of
10 L/tnin.
Background subtraction binding sensorgrams were used for analyzing the rate
constant of association (k8) and dissociation (kd), and the equilibrium
dissociation constant
KD. The resulting data sets were fitted with a 1:1 Langmuir Binding Model
using the Biacore
T200 evaluation software (version 2.0). Table 3 summarizes the affinities for
the anti-human
TIGIT antibodies to human TIGIT-His protein.
Table 3: Measurement of Affinity for anti-Human TIGIT Antibodies to human
TIGIT-
His protein and TIGIT-Fe protein using BlAcore.
BIAcore
KD (human TIGIT-his)
Clone (PM)
14D7 (mIgGl/K) 4.6
26B10 (mIgGI/K) 140
Comparator MBSA43 (inIgG1) 36.3
In Vitro T-cell Activity Assay for Antagonistic Anti-hTIGIT Antibodies
One assay we developed to characterize the functional consequence of blocking
human TIGIT receptor utilized Jurkat cells, an immortalized line of human T
lymphocyte
cells (clone, E6-1: ATCC TIB-152), engineered to over-express human TIGIT
(hTIGIT-
Jurkat) which were co-cultured with THP-1 cells, a human monocytic cell line
in the
presence or absence of one of the TIGIT ligands, CD155 and CD112. hTIGIT-
Jurkat cells
-88-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
co-cultured with THP-1 cells and stimulated with plate-bound anti-CD3 mAb
produce IL-2,
but when TIGIT ligand (CD155 or CD112) is added to the co-culture, IL-2 levels
were
reduced in a ligand-dependent manner. Treatment with antibodies that blocked
the CD155-
or CD112-TIGIT interaction, such as a commercially available anti-hTIGIT Ab,
clone
MBSA43 (eBioscience Cat# 12-9500-42; 10 pg/m1) rescues IL-2 to a level equal
to when
to activated hTIGIT Jurkat cells are cultured with THP-1 in the absence of
CD155 or CD112
(Figure 3).
96-well flat-bottom plates were coated with mouse anti-human CD3 antibody
(lug/m1
in PBS; Clone HIT3a; BD Pharmingen Cat# 555336) overnight at 4 C. The next
day,
hTIGIT-Jurkat cells (50,000) were plated in the pre-coated plates and pre-
incubated for 30-60
minutes with mAb at varying concentrations. THP-1 cells (50,000) were added to
the culture
followed by either CD155-Fc (ECD of human CD155 fused to human Fe; 1.0 pg/m1)
or
CD112-Fc (ECD of human CD112 fused to human Fc; 0.5 ttglin1). After incubation
for 18-
24h at 37 C and 5.0% CO2, IL-2 levels were assessed in culture supernatants by
Meso Scale
(Human IL-2 Tissue Culture MESO Kit: Cat#K151AHB-2).
As shown in Figure 3, a titration of the anti-hTIGIT clones 14D7 and 26B10
from 30
Itglinl down to 0.04 ig/m1 gave EC5Os of 0.17 pg/m1 and 0.19 pg/ml,
respectively, as
compared to MBSA43 at 0.23 tg/m1 using this assay.
Example 3
Humanization of Antibodies
The 14D7 and 26B10 mouse antibodies were humanized using methods described in
the specification. From antibody 14D7, the following humanized variable heavy
chains were
constructed: SEQ ID NOs: 9-12; and the following humanized variable light
chains were
constructed: SEQ ID NOs: 13-16. From mouse antibody 26B10, the following
humanized
variable heavy chains were constructed: SEQ ID NOs: 25-28; and the following
humanized
variable light chains were constructed: SEQ ID Nos: 29-32.
Example 4
Epitope Mapping of human TIGIT antibody by Hydrogen Deuterium Exchange Mass
Spectrometry
The contact areas between anti-TIGIT antibody 14D7 and human TIGIT were
determined by use of hydrogen deuterium exchange mass spectrometry (HDX-MS)
analysis.
-89-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
HDX-MS measures the exchange of deuterium with hydrogen into the amide
backbone of the
protein. One factor influencing the exchange rate is the hydrogen's exposure
to solvent.
Comparison of the exchange levels in the antigen when the antibody is bound
can identify
regions of the protein where the antibody is binding.
1.0 Materials
= hTIGIT-His (SEQ ID NO:35) ¨ Comprising the extracellular domain of human
TIGIT
(residues 25-145 of SEQ ID NO:33) and a histidine tag.
= Mouse anti-human TIGIT 14D7 antibody OW 26AHB) (Mouse x mAb
(MEB125.14D7.C1.C1) IgG1 / Kappa (HY)) (comprises the heavy chain variable
region of SEQ ID NO:6 and the light chain variable region of SEQ ID NO:?)
=
Liquid Chromatography-Mass Spectrometry
The mass spectrometer was a Thermo Scientific Orbitrap-Elite. For the
measurement of
deuterium labeled samples, the mass spectrometer was set to acquire one full
scan MS data in
the orbitrap at 120,000 resolving power, a target ion count of 1E6, a maximum
ion injection
time of 500 milliseconds and two microscans. For the acquisition of MS/MS data
for peptide
identifications, the mass spectrometer was set to acquire one full scan
spectrum at 120,000
resolving power followed by ten data-dependent MS/MS spectra in the ion trap.
The liquid chromatography system was a Waters nanoAcquity for the analytical
column
gradient and a Waters 515 isocratic pump for the sample digestion and loading.
For sample
digestion and loading, the buffer used was 2% acetonitrile and 0.05%
trifluoroacetic acid at a
flow rate of 80 gUminute. For the analytical gradient, the buffers were Buffer
A) 0.1%
formic acid in water and Buffer B) 0.1% formic acid in acetonitrile.
The gradient was at 40 IA/minute from 2% B to 36% B in 10 minutes, followed by
a
wash of 80% B for two minutes and a re-equilibration at 2% B for three
minutes. The
column was then washed by cycling the gradient between 2% and 80% B, three
times with
one minute at each step, followed by a final equilibration at 2% B for five
minutes. The
trapping column was a Waters Vanguard CSH C18 1.7 gm Guard Column and the
analytical
column was a Waters CSH C18, 1.7 gm 1x50 mm column.
Sample handling for the deuterium labeling was done by a Leaptec H/D-X PAL
system.
The labeling sample tray was set to a temperature of 25 C, the quenching tray
was set to
1.5 C and the trap and analytical column chamber was set to 1.5 C. The
immobilized pepsin
-90-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
column (Enzymate BEH Pepsin, Waters corporation) was kept outside the column
chamber at
room temperature.
Deuterium Labeling
Human TIGIT-His was mixed with the antibody 14D7 to final concentrations of 30
faM
for human TIGIT-his and 15 MM for 14D7. An unbound control was prepared by
incubating
human TIGIT-his in 50 inM sodium acetate pH 5.5. The antibody bound sample and
the
unbound control were incubated at room temperature for one hour before
beginning the
labeling experiment.
To deuterium label the samples, two !IL of sample was mixed with 25 !IL of PBS
in
deuterium oxide pH 7.6. Labeling time points were 30, 300, 1500, 4500 and 9000
seconds.
After the time point, 25 pL of the labeling mixture was added to 35 AL of cold
quench buffer
(8 M Urea, 100 mM TCEP). The quenched sample was incubated at 1.5 C for one
minute.
50 ML was then injected into the column cooling chamber where the sample was
passed over
the pepsin column and the resulting peptides loaded onto the trapping column.
After two
minutes, a valve switch took the pepsin column out of line and the trap was
washed at
additional 90 seconds. The trap was then switched in-line with the analytical
column and the
analytical gradient and the mass spectrometer data acquisition was started.
Each time point
was acquired in triplicate in randomized order.
A fully deuterated sample was generated by incubating 2 tit of hTIGIT (60
pmol/ ML)
with 108 ML of deuterated denaturing buffer (4 M Urea, 100 mM TCEP, 0.01% DDM
in
99.5% deuterium oxide). The sample was incubated at room temperature
overnight. 50 1.11,
was then directly injected into the column chamber and the data acquired as
before.
Data Analysis
LC-MS/MS data was acquired of an unlabeled sample and database searched to
verify
successful digestion of the proteins and to generate a list of peptides from
the pepsin
digestion. Database search was done using Proteome Discoverer 1.4 and the
SEQUEST HT
search algorithm (ThermoFisher Scientific). The protein database used was the
human
TTGIT-His and anti-human -MIT antibody sequences concatenated to the yeast
S'accharomyees eerevisiae Uniprot (5/20/13) database.
MS data from the deuterium labeling experiment was processed by HDExaminer
(version 1.3, Sierra Analytics). The mass and retention time selected by the
software for each
peptide was verified manually.
-91-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Results
The human TIGIT peptides protected by the 14D7 antibody are illustrated in the

heatmap (Figure 4) and correspond to amino acid residues 24-41
(SSTTAQVNWEQQDQL,
SEQ ID NO:113), 44-46 (ICN), 85-93 (IYHTYPDGT, SEQ ID NO:114) and 96-100
(GRIFL,
1.0 SEQ ID NO:115) of the human TIGIT shown in (SEQ ID NO:33).
All references cited herein are incorporated by reference to the same extent
as if each
individual publication, database entry (e.g. Genbank sequences or GeneID
entries), patent
application, or patent, was specifically and individually indicated to be
incorporated by
reference. This statement of incorporation by reference is intended by
Applicants, pursuant
to 37 C.F.R. 1.57(b)(1), to relate to each and every individual publication,
database entry
(e.g. Genbank sequences or GeneID entries), patent application, or patent,
each of which is
clearly identified in compliance with 37 C.F.R. 1.57(b)(2), even if such
citation is not
immediately adjacent to a dedicated statement of incorporation by reference.
The inclusion
of dedicated statements of incorporation by reference, if any, within the
specification does
not in any way weaken this general statement of incorporation by reference.
Citation of the
references herein is not intended as an admission that the reference is
pertinent prior art, nor
does it constitute any admission as to the contents or date of these
publications or documents.
The present invention is not to be limited in scope by the specific
embodiments
described herein. Indeed, various modifications of the invention in addition
to those
described herein will become apparent to those skilled in the art from the
foregoing
description and the accompanying figures. Such modifications are intended to
fall within the
scope of the appended claims.
The foregoing written specification is considered to be sufficient to enable
one skilled
in the art to practice the invention. Various modifications of the invention
in addition to
those shown and described herein will become apparent to those skilled in the
art from the
foregoing description and fall within the scope of the appended claims.
Table 4: Sequence Information
Description SEQ SEQUENCE
ID
NO:
14D7 H - CDR1 1 GAWMD
14D7 H - CDP2 2 EIRTKVNNHATNYGESVKG
141)7 H - CDR3 3 ALYDGFYFDY
-92-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
14D7 L - CDR1 4 SASSSVSSGYLY
14D7 L - CDR2 5 GTSTLAS
14D7 L - CDR3 6 HQWSSFPYT
1407 VH 7
EVKLEESGGGLVQPGGSMKLSCVASGFTFSGAWMDWVRQSPEKGLEWVAEIRTKVNNHATNY
PARENTAL GESVKGRFTISRDDSKSSVYLQMNNLRAEDSGIYYCRGALYDGFYFDYWGQGTTLTVSS
14D7 VL 8
QIVLTQSPAIMSASPGEKVNLTCSASSSVSSGYLYWYQQKPGSSPKLWIYGTSTLASGVPAR
PARENTAL FSGSGSGTSYSLT1SNMEAEDAASYFCHNSSFPYTFGGGTKLEMK
Hul4D7 VH 9 EVQLVESGGGLVQ PGGSL KLSCAASGFT FS GAX: X2DWVRQAPGKGLEWVAEI
RTKVNNHATN
humanized
YGESVKGRFTISRDX3SKX4X,VYLQX6X7X8LRAEDX9AVYYCRGALYX10XliFYFDYWGQGTL
consensus VTVSS
sequence )(1. W, A, R, N, D, Q, E, G, H, I, L, K, F, Pr 5, T, Y, V
X.-- M, V, L, I, G, A, S, T
= D, A, R, N, Q, E, G, H, I, L, K, F, Sr Tr Yr V
Xe S, N
= T, S
= M, L
X7= N, S
X8= 5, N
X9= T, S
X10= D, A, R, N, Q, E, G, H, I, L, K, F, P, 5, T, W, /, V
X11= G, A, R, N, D, Q, E, H, I, L. K, F, P, 5, T, W, /, V
HU14D7 VH1 10
EVQLVESGGGLVQPGGSLKLSCAASGFTESCAVVRQAPGKGLEWVAEIRTKVNNHAXNY
(Humanized VH GESVKGRFTISRDDSKSTVYLQMNSLRAED:LAVYYRGALYDGFYFDYWGQGTLVTVSS
chain)
Hu14D7 VH2 11
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVXGRFTISRDDSKSSVYLQMNSLRAEDTAVYYCRGALYDGFYFDYWGQGTLVTVSS
chain)
Hu14D7 VH3 12
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKNTVYLQMNSLRAEDTAVYYCRGALYDGFYFDYWGQGTLVTVSS
chain)
Hu14D7 VL 13
EIVLTQSPATLSLSPGERAX1LSCSABSSVSSGYLYWYQQKPGQAPX7LX2IYGTSTLASGX8P
humanized ARFSGSGSGTDYTLTISSX3EPEDX4AVYYCHQX5SSFPYTFGQGTKLEX6K
consensus Xl= T, S
Sequence X2= W, A, R, N, D, Q, E, G, H, I, L, K, Fr Pr 5, T, Y, V
X3= L, V, I
X4= F, V, L, I, T
X5= W, A, R, N, D, Q, E, G, H, I, L, K, Fr Pr 5, T, Y, V
X6= I, L
X7= K, R
X8= V, I
HU14D7V Li 14
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQWSSFPYTFGQGTKLEIK
chain)
Hu14D7V L2 15
E1VLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPRLW1YGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQWSSFPYTFGQGTKLEIK
chain)
Hu14D7V L3 16
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPRLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQWSSFPYTFGQGTKLEIK
chain)
26810 H - 17 EFTMH
CDR1
26810 H - 18 GLKPDNGGISYNQKFKG
CDR2
26810 H - 19 GAYYRYDADY
CDR3
26810 L - 20 KASQDVKTAVA
CDR1
26810 L - 21 SASYRNT
CDR2
26810 L - 22 QQHYSTPFT
CDR3
26810 VH 23
EVQLQQSGPELVKPGASVKISCKTSGYTFTEFTMHWVKQSHGKSLEWIGGLKPDNGGISYNQ
PARENTAL KFKGRATLAVDKSSNTAYMELRSLTSEDSAVYYCARGAYYRYDADYWGQGTTLTVSS
26810 VL 24
DIVITQSHKFMSTSVGDRVSITCKASQDVKTAVAWYQUSGQSPKLLIYSASYRNTGVPDRF
PARENTAL TGSGSGTDFTFTIDSVQAEDLAVYFCQQHYSTPFTFGTGTKLELK
-93-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
26810 VH 25 EVQLVQSGAEVKKPGASVKISC3GMILWVX3QAPGKGLEWIGGLKPDX4K5GIS
HUMANIZED YNQKFKGRATLTVDX6STX7TAYSS5U)X9AVYYCARGAYYRYX10X11DYWGQGTLVT
CONSESUS VSS
Sequence X1 T, V
X2= M, V, L, 1, G, A, S, T
X3= K, R
X4= N, P, R, D, Q, E, G, H, 1, L, K, F, P, S, T, W, 1, V
X5= G, P, R, N, D, Q, E, H, 1, L, K, F, P, S, T, W, 1, V
X6= k, t, d, s
X7= N, S
X8= M, V, L, 1, G, A, S, T
Xy= T, S
D, A, R, N, Q, E, G, H, I, L, K, F, P, S, T, W, Y, V
X11 R, N, D, Q, E, G, H, I, L, K, F, P, S, T, W, Y, V, M
Hu26810 VH1 26
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDNGGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)
Hu26810 VH2 27
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDNGG1SYNQ
(Humanized VH KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)
Hu26810 VH3 --28
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDNGGISYNQ
(Humanized VH KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)
26810 Vi 29
DIQLTQSPSSLSASVGDPVTITCKASQDVKTAVAWYQQKPGKAPKLLIYSASYRX1X7GVP
HUMANIZED X3RFSGSGSGTDFTX4TISSLQPEDFATYYCQQHYSTPFTFGQGTKLEIK
CONSESUS Xl= N, Q, D, E
sequence X2= T, S, A
X3= D, S
X4. F, L
Hu26810 VL1 30
DIQLTQSPSSLSASVGDRVTITCKASQDVKTAVAWYQQKPGKAPKLLIYSASYRNTGVPDRF
(Humanized VL SGSGSGTDFTFTISSLQPEDFATYYCQQHYSTPFTFGQGTKLEIK
chain)
Hu26810 VL2 31
DIQLTQSPSSLSASVGDRVTITCKASOVKTAVAWYQUPGKAPKLLIYSASYRNTGVPSRF
(Humanized VL SGSGSGTDFTFTISSLQPEDFATYYCQQHYSTPFTFGQGTKLEIK
chain)
Hu26810 VL3 32
DIQLTQSPSSLSASVGDRVTITCKASQDVKTAYAWYQQKPGKAPKLLIYSASYRNTGVPSRF
(Humanized VI SGSGSGTDFTLTISSLQPEDFATYYCQQHYSTPFTFGQGTKLEIK
chain)
Human 33 mrwcilliwa qgirgaplas gmmtgtiett gnisaekggs iilqchlsst
TIGIT(24 taqvtgvnwe qqdqllaicn adlgwhisps fkdrvapgpg 1g1tIgsltv
amino acid ndLgevfciy htypdgtyLg riflevless vaehgarfqi pllgamaatl
leader in vvictavivv valtrkkkal rihsvegdlr rksagqeews psapsppgsc
italics & vgaeaapagl cgeqrgedca elhdyfnvls yrslgncsff tetg
extracellular
domain
underlined)
Cyno / Rhesus 34 mrwelfliwa gglrgaplas gmmtgtiett gnisakkggs vilgchlsst
TIG1T magvtgvnwe qhdhsllair naelgwhiyp afkdrvapgp glgltlgslt
mndtgeyfct yhtypdgtyr griflevles svaehsarfq ipllgamamm
lvviciaviv vvvlarkkks lrihsvesgl grkstggeeg ipsapsppgs
cvgaeaapag legegggddc aelhdyfnvl syrslgscsf ftetg
hTIGIT-HIS 35 gtiett gnisaekggs iilqchlsst taqvtqvnwe qqdqllaicn
adlgwhisps
(extra- fkdrvapgpg 1g1tIgsltv ndtgeyfciy htypdgtytg riflevless
cellular vaehgarfqi pllga hhhhhhhhhggq
domain of
human TIGIT
fused to His
tag)
Pembrolizumab 36
QVQLVQSGVEVKKPGASVKVSCKASGYTFTN"r/AYV.VRQAi'GQGLEVIMGGINPSNGGTNFNE
Heavy chain
KFKNRVTLTTDSSTTTAYMELKSLQFDDTAVYYCARRDYRFDMGFDYWGQGTTVTVSSASTK
GPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS
SVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPK
DTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLH
QDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGF
YPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHN
HYTQKSLSLSLGK
-94-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
Pembrolizumab 37 EIVLTQSPATLSLSPGERATLSCRASKGVSTSGYSYLHWYQUPGQAPRLLIYLASYLESGV
Light chain
PAPFSGSGSGTDFTLTISSLEPEDFAVYYCQHSPDLPLTFGGGTKVEIKRTVAAPSVFIFPP
SDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALOGNSQESVTEQDSKDSTYSLSSTLTLS
KADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
Nivolwuab 38 QVQLVESGGGVVQPGRSLRLDCKASG1TFSNSGMHWVRQAPGKGLEWVAVIWYDGSKRYYAD
heavy chain
SVKGRFTISRDNSKNTLFLQMNSLRAEDTAVYYCATNDDYWGQGTLVTVSSASTKGPSVFPL
APCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFENVLQSSGLYSLSSVVTVPS
SSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKYKDTLMISR
TPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGK
EYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDLAV
EWESNWPENNYKTTPPVLDSDGSFFLYSRLTVDKSRKEGNVFSCSVMHEALHNHYTQKSL
SLSLGK
Nivolumab 39 EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQUPGQAPRLLIYDASNRATGIPARF
light chain
SGSGSGTDFTLTISSLEPEDFAVYYCQQSSNWPRTFGQGTKVEIKRTVAAPSVFIFPPSDEQ
LKSGTASVVCLLNNFYPREAKVQWKVDNALOGNSQESVTEUSKDSTYSLSSTLTLSKADY
EKHKVYACEVTHQGLSSPVTKSFNRGEC
Leader 40 MEWSWVFLFFLSVTTGVHS
sequence
heavy chains
Leader 41 MSVPTQVLGLLLLWLTDARC
sequence
light chains
Heavy chain 42
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGAITSGVHTFPAVLOSGL
constant
YSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFP
domain-
PKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVL
IgG4
TVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCL
S228P
VKGFYPSDIAVEWESNWPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHE
ALHNHYTQKSLSLSLGK
Kappa light 43
VAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDS
chain TYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
constant
domain
Heavy chain 44
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL
constant
YSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPA2ELLGGPSVF
domain-
LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVV
IgG1
SVLTVLHQDWLNGKEYKCKVSNKALRAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSL
TCLVKGFYPSDIAVEWESNGUENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV
MHEAIHNHYTQKSLSLSPGK
1407 H - CDR3 45 ALYEGFYFDY
(D104E)
14D7 H - CDR3 46 ALYDAFYFDY
(G105A)
14D7 H - CDR3 47 ALYDSFYFDY
(G1055)
Hu14D7 VH1 48
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKSTVYLQMNSLRAEDTAVYYCRGALYEGFYFDYWGQGTLVTVSS
chain)
(D104E)
Hu1407 VH1 49
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAE1RTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKSTVYLQMNSLRAEDTAVYYCRGALYDAFYFDYWGQGTLVTVSS
chain)
(G105A)
Hu14D7 VH1 50
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKSTVYLQMNSLRAEDTAVYYCRGALYDSFYFDYWGQGTLVTVSS
chain)
(G105S)
Hul4D7 VH2 51
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKSSVYLQMNSLRAEDTAVYYCRGALYDEFYFDYWGQGTLVTVSS
chain)
(D104E)
Hul4D7 VH2 52
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVKAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH CESVKCith-
TISRDDSKSSVYLQMNSLRAEDTAVYYCRCALYDAFYFDYWGQGTLVTVSS
chain)
-95-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
(Glow
Hul4D7 VH2 53
EVQLVESGGGLVUGGSLKLSCANSGFTFSGAWADWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKSSVYLQMNSLRAEDTAVYYCRGALYDSFYFDYWGQGTLVTVSS
chain)
(G105S)
Hu14D7 VH3 54
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAALV.VRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKNTVYLQMNSLRAEDYAVYYCRGALYEGFYFDYWGQGTLVTVSS
chain)
(D104E)
Hu14D7 VH3 55
EVQLVESGGGLVQPGGSLKLSCAASGFTFSGAWMDWVRQAPGKGLEWVAEIRTKVNNHATNY
(Humanized VH GESVKGRFTISRDDSKNTVYLQMNSLRAEDTAVYYCRGALYDAFYFDYWGQGTLVTVSS
chain)
(G1 05A)
14u14D7 VH3 56 EVQLVESGGGLVQPGGSLKLSCALS:3inS.CAI;L-
IDWVRQAPGKGLEWVAEIRTICVNNHATNY
(Humanized VH GESVKGRFTISRDDSKNTVYLQMNSLRAEDTAVYYCRGALYDSFYFDYWGQGTLVTVSS
chain)
(G105S)
14D7 L - CDP3 57 HQASSFPYT
(W92A)
14D7 L - 58 HQDSSFPYT
CDR3(W92D)
14D7 L - 59 HQESSFPYT
CDR3(W92E)
14D7 L - 60 HQFSSFPYT
CDR3(W92F)
14D7 L - 61 HQGSSFPYT
CDR3(W92G)
14D7 L - CDR3 62 HQHSSFPYT
(W92H)
Hui4D7V Li 63
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYWKPCQA2KLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDEAVYYCHQASSFPYTFGQGTLEIK
chain) (W92A)
Hu14D7V Li 64
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQch.P:_:QAKLWISTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQDSSFPYTFGQGTKLEIK
chain) (W92D)
Hul4D7V Li 65
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQESSFPYTFGQGTKLEIK
chain) (W92E)
Hul4D7V Li 66
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQFSSFPYTFGQGTKLEIK
chain) (W92F)
Hu14D7V Li 67
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTSTLASGVPAR
(Humanized VI FSGSGSGTDYTLTISSLEPEDFAVYYCHQGSSFPYTFGQGTKLEIK
chain) (W92G)
Hu14D7V Li 68
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPKLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQHSSFPYTFGQGTKLEIK
chain) (W92H) 1
Hul4D7V L2 69
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPPLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQASSFPYTFGQGTKLEIK
chain) (W92A)
Hul4D7V L2 70
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYWKPGQAPPLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQDSSFPYTFGQGTKLEIK
chain) (W92D)
Hu14D7V L2 71
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPRLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQESSFPYTFGQGTKLEIK
chain) (W92E)
Hu14D7V L2 72
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYWKPGQAPPLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQFSSFPYTFGQGTKLEIK
chain) (W92F)
Hu14D7V L2 73
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYWKPGQAPPLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQGSSFPYTFGQGTKLEIK
chain) (W92G)
Hu14D7V L2 74
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYWKPGQAPPLWIYGTSTLASGVPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQHSSLPYTFGQGTKLEIK
chain) (W92H)
Hu14D7V L3 75
EIVLTQSPATLSLSPGERATLSCSASSSVSLYV;YQQKPGQAPPLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQASSFPYTFGQGTKLEIK
-96-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
chain) (W92A)
Hu14D7V L3 76
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYi,YWYW,KPGQAPkJ,w.CiGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQDSSFPYTFGQGTKLEIK
chain) (W92D) __
Hu14D7V L3 77
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYi:YWYWKPGQAPRLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQESSFPYTFGQGTKLEIK
chain) (W92E)
Hu14D7V L3 78
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPRLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCKFSSYTFGQGTKLEIK
chain) (W92H) __
Hul4D7V L3 79
EIVLTQSPATLSLSPGERATLSCSASSSVSS_YWYWKPGQAPRLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQGSSEYTFGQGTKLEIK
chain) (W92G)
Hu14D7V L3 80
EIVLTQSPATLSLSPGERATLSCSASSSVSSGYLYWYQQKPGQAPRLWIYGTSTLASGIPAR
(Humanized VL FSGSGSGTDYTLTISSLEPEDFAVYYCHQHSSFPYTFGQGTKLEIK
chain) (W92H)
26810 H - 81 GLKPDQGGISYNQKFKG
CDR2 (N55Q)
26810 H - 82 GLKPDDGGISYNQKFKG
CDR2(N55D)
26810 H - 83 GLKPDNAGISYNQKFKG
CDR2(N56A)
26810 H - 84 GLKPDTGGISYNQKFKG
CDR2(N55T)
26810 H - 85 GLKPDSGGISYNQKFKG
CDR2(N55S)
26810 H - 86 GLKPDGGGISYNQKFKG
CDR2(N55G)
26810 H - 87 GLKPDNSGISYNQKFKG
CDR2 (G56S)
26810 H - 88 GLKPDNTGISYNQKFKG
CDR2(G56T)
Hu26810 Viii 89
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDQGGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55Q)
Hu26810 Viii 90
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDDGGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55D)
Hu26810 VH1 91
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDNAGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYPYDADYWGQGTLVTVSS
chain)(G56A)
Hu26810 VH1 92 EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGC_FLT
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55T)
Hu26810 VH1 93
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDSGGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55S)
Hu26810 VH1 94
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDGGGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55G)
Hu26810 Viii 95
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDNSGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYPYDADYWGQGTLVTVSS
chain)(G56S)
Hu26810 VH1 96
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVKQAPGKGLEWIGGLKPDNTGISYNQ
(Humanized VH KFKGRATLTVDKSTNTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56T)
Hu26810 VH2 97
EVQLVQSGA8VKKPGASVKISCKTSGY1FTEFTMHWVRQAPGKGLEWIGGLKPDQGGISYNQ
(Humanized VH KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55Q)
Hu26810 VH2 98
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDDGGISYNQ
(Humanized VH KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55D)
Hu26810 Viii 99
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDNAGISYNQ
(Humanized VH KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56A)
Hu26810 VH2 100
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDTGGISYNQ
(Humanized VH KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
-97-

CA 02994555 2018-02-01
WO 2017/030823
PCT/US2016/046100
chain)(N55T)
Hu26B10 VH2 101
EVQLVOGAEVKKPGASVK1SCKTSGYTFTEFTAHWVRQAPGKCLEW1GCLKPDSGGISYNQ
(Humanized VH
KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55S)
Hu26810 VH2 102
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDGGGISYNQ
(Humanized VH
KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVPVSS
chain)(N55G)
Hu26B10 VH2 103
EVQLVQSGAEVKKPGASVKISCKTSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDNSGISYNQ
(Humanized VH
KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYPYDADYWGQGTLVTVSS
chain)(G56S)
1iu26810 VH2 104 EVQLVQSGAEVKKPGASVKISCKTSGYTi 1F
(Humanized VH
KFKGRATLTVDKSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56T)
Hu26B10 VH3 105
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDQGGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55Q)
Hu26810 VH3 106
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDDGGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55D)
Hu26810 VH3 107 -EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHW-
VnAPGKGLEWIGGLKPDNAG1SYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56A)
Hu26810 VH3 108
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDTGGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55T)
Hu26810 VH3 109
EVQLVQSGAEVKKPGASVK1SCKVSGY1ETEETMHWVRQAPGKGLEW1GGLKPDSGGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55S)
Hu26810 VH3 110
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDGGGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(N55G)
Hu26810 VH3 111
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKPDNSG1SYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56S)
Hu26810 VH3 112
EVQLVQSGAEVKKPGASVKISCKVSGYTFTEFTMHWVRQAPGKGLEWIGGLKETNTGISYNQ
(Humanized VH
KFKGRATLTVDTSTSTAYMELSSLRSEDTAVYYCARGAYYRYDADYWGQGTLVTVSS
chain)(G56T)
hTIGIT 113 SSTTAQVNWEQQDQL
epitope (24-
41)
hTIGIT 114 IYHTYPDGT
epitope (85-
93)
hTIG1T 115 GRIFL
epitope (96-
100)
-98-

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2024-07-02
(86) PCT Filing Date 2016-08-09
(87) PCT Publication Date 2017-02-23
(85) National Entry 2018-02-01
Examination Requested 2021-05-27

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $210.51 was received on 2023-12-15


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2025-08-11 $100.00
Next Payment if standard fee 2025-08-11 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Registration of a document - section 124 $100.00 2018-02-01
Application Fee $400.00 2018-02-01
Maintenance Fee - Application - New Act 2 2018-08-09 $100.00 2018-02-01
Maintenance Fee - Application - New Act 3 2019-08-09 $100.00 2019-07-17
Maintenance Fee - Application - New Act 4 2020-08-10 $100.00 2020-07-13
Request for Examination 2021-08-09 $816.00 2021-05-27
Maintenance Fee - Application - New Act 5 2021-08-09 $204.00 2021-07-13
Maintenance Fee - Application - New Act 6 2022-08-09 $203.59 2022-07-12
Registration of a document - section 124 $100.00 2022-10-12
Extension of Time 2022-10-24 $203.59 2022-10-24
Maintenance Fee - Application - New Act 7 2023-08-09 $210.51 2023-07-11
Maintenance Fee - Application - New Act 8 2024-08-09 $210.51 2023-12-15
Final Fee $416.00 2024-05-21
Final Fee - for each page in excess of 100 pages 2024-05-21 $56.00 2024-05-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MERCK SHARP & DOHME LLC
Past Owners on Record
MERCK SHARP & DOHME CORP.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Request for Examination 2021-05-27 3 73
Claims 2018-02-02 5 268
Examiner Requisition 2022-06-22 4 252
Extension of Time 2022-10-24 3 77
Acknowledgement of Extension of Time 2022-11-28 2 221
Amendment 2022-12-21 22 1,662
Claims 2022-12-21 6 334
Description 2022-12-21 98 10,314
Abstract 2018-02-01 2 73
Claims 2018-02-01 6 431
Drawings 2018-02-01 4 61
Description 2018-02-01 98 9,219
International Search Report 2018-02-01 5 224
Declaration 2018-02-01 2 48
National Entry Request 2018-02-01 10 406
Voluntary Amendment 2018-02-01 6 294
Prosecution/Amendment 2018-02-02 2 51
Representative Drawing 2018-03-26 1 6
Cover Page 2018-03-26 1 33
Final Fee 2024-05-21 3 88
Representative Drawing 2024-05-30 1 8
Interview Record Registered (Action) 2023-10-24 1 16
Amendment 2023-11-08 10 337
Claims 2023-11-08 5 326

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :